1
|
Akuta T, Kurosawa Y, Tomioka Y, Arakawa T. Impact of arginine addition on protein concentration via ultrafiltration. Protein Expr Purif 2025; 231:106701. [PMID: 40112936 DOI: 10.1016/j.pep.2025.106701] [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: 01/20/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
In this study, we examined the effects of arginine (L-ArgHCl) on ultrafiltration performance, a process with practical significance for not only research but also pharmaceutical applications. Specifically, we assessed the yield and filtration rate of ultrafiltration using rabbit and goat polyclonal antibodies (neutral to basic isoelectric points) as well as model proteins, BSA (acidic) and lysozyme (basic). When a 1 mg/mL protein solution was concentrated approximately 10-fold using a standard commercially available centrifugal ultrafiltration device, the addition of L-ArgHCl significantly improved yield at near-neutral buffer pH. The observed order of improvement was: 20 mM L-ArgHCl >100 mM L-ArgHCl ≈0.5 M NaCl > no addition. A similar trend was observed with BSA, whereas lysozyme achieved slightly higher yields at 100 mM L-ArgHCl. In a 40-fold concentration of rabbit polyclonal antibody from 1 mg/mL, 20 mM L-ArgHCl enhanced yield at pH 6 and 7, but had minimal or no effect at pH 7.5. Notably, at pH 8, high yields were achieved without arginine. L-ArgHCl also accelerated the concentration rate at all pH levels, with greater enhancements observed at higher arginine concentrations. These findings suggest that L-ArgHCl mitigates protein precipitation and solubility loss by reducing protein-protein interactions and nonspecific binding to the ultrafiltration membrane. At pH 8, the increased surface charge of the antibody reduced hydrophobicity, further improving solubility. In summary, the addition of L-ArgHCl, particularly near pH 7, effectively enhanced ultrafiltration performance. This provides a practical strategy for improving protein concentration processes.
Collapse
Affiliation(s)
- Teruo Akuta
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd., Aza-Asayama, Kamitezuna, Takahagi, Ibraki, 3333-26, Japan.
| | - Yasunori Kurosawa
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd., Aza-Asayama, Kamitezuna, Takahagi, Ibraki, 3333-26, Japan
| | - Yui Tomioka
- Research and Development Division, Kyokuto Pharmaceutical Industrial Co., Ltd., Aza-Asayama, Kamitezuna, Takahagi, Ibraki, 3333-26, Japan
| | - Tsutomu Arakawa
- Alliance Protein Laboratories, 13380 Pantera Rd, San Diego, CA, 92130, USA.
| |
Collapse
|
2
|
Kasahara J, Furuki T, Aikawa S, Ueda H, Shiraki K. Polyphosphate as a novel aggregation suppressor of gamma globulin. J Pharm Sci 2025:103818. [PMID: 40349926 DOI: 10.1016/j.xphs.2025.103818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 04/26/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
The aggregation of gamma globulin poses a significant challenge in maintaining the quality of biopharmaceutical products. This study aimed to develop a novel approach to prevent gamma globulin aggregation using polyphosphates (PolyPs), linear polymers comprising 14 to 130 phosphate units. The addition of PolyPs effectively suppressed the formation of subvisible particles (SVPs) in the micrometer-sized fraction of bovine gamma globulin (BGG) during storage at 40°C, as observed through flow imaging. Furthermore, PolyPs mitigated the decrease in soluble protein concentration under these conditions. Mass photometry and isothermal titration calorimetry revealed that PolyPs spontaneously form complexes with BGG. The negative zeta potential and positive B22 and kDiff values suggested that the BGG-PolyP complexes were stabilized by electrostatic repulsion. Importantly, far-UV circular dichroism confirmed that the secondary structure of BGG remained unaffected by complexation with PolyPs. Notably, arginine-a commonly used aggregation suppressor-failed to prevent the formation of SVPs in BGG under similar conditions. This study demonstrates the potential of biocompatible and stable PolyPs as a novel additive for inhibiting gamma globulin aggregation, offering a promising alternative to conventional approaches.
Collapse
Affiliation(s)
- Junpei Kasahara
- Faculty of Pure and Applied Sciences, University of Tsukuba, Ibaraki 305-8573, Japan; Vaccine Manufacturing Technology Research Laboratory, Shionogi Co., Ltd., Osaka 561-0825, Japan.
| | - Tomohiro Furuki
- Faculty of Pure and Applied Sciences, University of Tsukuba, Ibaraki 305-8573, Japan
| | - Shohei Aikawa
- Vaccine Manufacturing Technology Research Laboratory, Shionogi Co., Ltd., Osaka 561-0825, Japan
| | - Hiroshi Ueda
- Analysis and Evaluation Laboratory, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Kentaro Shiraki
- Faculty of Pure and Applied Sciences, University of Tsukuba, Ibaraki 305-8573, Japan.
| |
Collapse
|
3
|
Yadav J, Uddin S, Civati F, Ma W, Liebminger A, Teschner W, André G, Trout BL, Braatz RD, Myerson AS. Developing ultra-high concentration formulations of human immune globulins for subcutaneous injectables. J Pharm Sci 2025; 114:1605-1614. [PMID: 39909221 DOI: 10.1016/j.xphs.2025.01.028] [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: 09/24/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
This work describes the first development of high-concentration suspension formulations of human immune globulin. Colloidal-level dispersions of immune globulin were achieved by suspending a spray dried solid powder of protein in a protein solution made saturated by the addition of pharmaceutical excipients. The spray drying process was used to generate ∼90 % of particles below 20μ. The monomer and aggregates content of immunoglobulin were found to be 93 % and 0.3 %, respectively. The injection forces for the colloidal suspensions were characterized using a dynamic compression test. The concentrations of 300, 380, and 400 mg/mL formulations were injected at 3.8 N, 10 N, and 16.5 N of maximum injection forces, respectively, when a 24-gauge needle was used. The viscosity of a 300 mg/mL suspension was 128 cP. The viscosity of a 380 mg/mL suspension was 284 cP, and the viscosity was higher for the 400 mg/mL formulation; however, injectability was not an issue, which remains rare for non-Newtonian, shear-thickening systems. It is acknowledged that the 400 mg/mL suspension formulation remained relatively challenging as compared to other suspensions for injection because of its very high viscosity, and significant force was required to inject it. We show that where ultra-high-concentration immune globulin is being developed within reasonable constraints of pharmaceutical regulation, with an injectability parameter, formulations might make their way to the clinic when viscosity could say otherwise. However, further work should be conducted to assess chemical stability (using methods such as mass spectrometry) along with forced degradation studies prior any clinical use.
Collapse
Affiliation(s)
- Jayprakash Yadav
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, United States
| | - Shihab Uddin
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, United States
| | - Francesco Civati
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, United States
| | - Wenchuan Ma
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, United States
| | - Andreas Liebminger
- Takeda Pharmaceuticals, Process Development, Plasma-derived Therapies R&D, Industriestraße 72, A-1220 Vienna, Austria
| | - Wolfgang Teschner
- Takeda Pharmaceuticals, Process Development, Plasma-derived Therapies R&D, Industriestraße 72, A-1220 Vienna, Austria
| | - Guillaume André
- Takeda Pharmaceuticals, Process Development, Plasma-derived Therapies R&D, Industriestraße 72, A-1220 Vienna, Austria
| | - Bernhardt L Trout
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, United States
| | - Richard D Braatz
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, United States
| | - Allan S Myerson
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139-4307, United States.
| |
Collapse
|
4
|
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
|
5
|
Armstrong GB, Shah V, Sanches P, Patel M, Casey R, Jamieson C, Burley GA, Lewis W, Rattray Z. A framework for the biophysical screening of antibody mutations targeting solvent-accessible hydrophobic and electrostatic patches for enhanced viscosity profiles. Comput Struct Biotechnol J 2024; 23:2345-2357. [PMID: 38867721 PMCID: PMC11167247 DOI: 10.1016/j.csbj.2024.05.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
The formulation of high-concentration monoclonal antibody (mAb) solutions in low dose volumes for autoinjector devices poses challenges in manufacturability and patient administration due to elevated solution viscosity. Often many therapeutically potent mAbs are discovered, but their commercial development is stalled by unfavourable developability challenges. In this work, we present a systematic experimental framework for the computational screening of molecular descriptors to guide the design of 24 mutants with modified viscosity profiles accompanied by experimental evaluation. Our experimental observations using a model anti-IL8 mAb and eight engineered mutant variants reveal that viscosity reduction is influenced by the location of hydrophobic interactions, while targeting positively charged patches significantly increases viscosity in comparison to wild-type anti-IL-8 mAb. We conclude that most predicted in silico physicochemical properties exhibit poor correlation with measured experimental parameters for antibodies with suboptimal developability characteristics, emphasizing the need for comprehensive case-by-case evaluation of mAbs. This framework combining molecular design and triage via computational predictions with experimental evaluation aids the agile and rational design of mAbs with tailored solution viscosities, ensuring improved manufacturability and patient convenience in self-administration scenarios.
Collapse
Affiliation(s)
- Georgina B. Armstrong
- Drug Substance Development, GlaxoSmithKline, Gunnels Wood Road, Stevenage, UK
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Vidhi Shah
- Large Molecule Discovery, GlaxoSmithKline, Gunnels Wood Road, Stevenage, UK
| | - Paula Sanches
- Drug Substance Development, GlaxoSmithKline, Gunnels Wood Road, Stevenage, UK
| | - Mitul Patel
- Drug Substance Development, GlaxoSmithKline, Gunnels Wood Road, Stevenage, UK
| | - Ricky Casey
- Drug Substance Development, GlaxoSmithKline, Gunnels Wood Road, Stevenage, UK
| | - Craig Jamieson
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Glenn A. Burley
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - William Lewis
- Drug Substance Development, GlaxoSmithKline, Gunnels Wood Road, Stevenage, UK
| | - Zahra Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
6
|
Esfahani IC, Tehrani NA, Ji S, Sun H. Simultaneous Protein Adsorption and Viscosity Measurement using Micropillar-Enhanced Acoustic Wave (μPAW) Device for Pharmaceutical Applications. J Pharm Sci 2024; 113:2715-2722. [PMID: 38857644 DOI: 10.1016/j.xphs.2024.06.001] [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: 01/27/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
At the early stages of drug development, the amount of drug materials is rather limited. In this case, viscosity measurement is often postponed to the later stages, where grams of proteins can be produced. Therefore, it is necessary to develop a viscometer capable of measuring the viscosity with high accuracy while requiring low sample volume. This study presents a novel viscosity measurement technique based on measuring the resonance frequency and motional resistance of a micropillar-enhanced acoustic wave (μPAW) device. The μPAW was developed by fabricating micropillars on the quartz crystal microbalance substrate in order to achieve ultra-high sensitivity, thanks to a unique coupling between the micropillar and the resonator. The experimental measurements demonstrated a nonlinear relationship between the density and viscosity of the fluid and the response of μPAW. A calibration correlation was developed using the response of μPAW in aqueous glycerol and sucrose solutions. The measurements were then extended using high-concentration BSA solutions as the model of protein solution. The main advantage of the μPAW device in this work over other viscometers is the ability to simultaneously measure solution viscosity and protein adsorption on the surface. This is a huge step forward in the development of sensing systems for the pharmaceutical industry, where real-time sensing of target biological proteins and measuring the viscosity of a solution is required.
Collapse
Affiliation(s)
| | - Nastaran A Tehrani
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, United States
| | - Siqi Ji
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, United States
| | - Hongwei Sun
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA 02115, United States.
| |
Collapse
|
7
|
Tanimoto S, Okumura H. Why Is Arginine the Only Amino Acid That Inhibits Polyglutamine Monomers from Taking on Toxic Conformations? ACS Chem Neurosci 2024; 15:2925-2935. [PMID: 39009034 PMCID: PMC11311134 DOI: 10.1021/acschemneuro.4c00276] [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: 05/10/2024] [Revised: 06/09/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Polyglutamine (polyQ) diseases are devastating neurodegenerative disorders characterized by abnormal expansion of glutamine repeats within specific proteins. The aggregation of polyQ proteins is a critical pathological hallmark of these diseases. Arginine was identified as a promising inhibitory compound because it prevents polyQ-protein monomers from forming intra- and intermolecular β-sheet structures and hinders polyQ proteins from aggregating to form oligomers. Such an aggregation inhibitory effect was not observed in other amino acids. However, the underlying molecular mechanism of the aggregation inhibition and the factors that differentiate arginine from other amino acids, in terms of the inhibition of the polyQ-protein aggregation, remain poorly understood. Here, we performed replica-permutation molecular dynamics simulations to elucidate the molecular mechanism by which arginine inhibits the formation of the intramolecular β-sheet structure of a polyQ monomer. We found that the intramolecular β-sheet structure with more than four β-bridges of the polyQ monomer with arginine is more unstable than without any ligand and with lysine. We also found that arginine has 1.6-2.1 times more contact with polyQ than lysine. In addition, we revealed that arginine forms more hydrogen bonds with the main chain of the polyQ monomer than lysine. More hydrogen bonds formed between arginine and polyQ inhibit polyQ from forming the long intramolecular β-sheet structure. It is known that intramolecular β-sheet structure enhances intermolecular β-sheet structure between proteins. These effects are thought to be the reason for the inhibition of polyQ aggregation. This study provides insights into the molecular events underlying arginine's inhibition of polyQ-protein aggregation.
Collapse
Affiliation(s)
- Shoichi Tanimoto
- Exploratory
Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki 444-8787, Aichi, Japan
| | - Hisashi Okumura
- Exploratory
Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki 444-8787, Aichi, Japan
- National
Institutes of Natural Sciences, Institute
for Molecular Science, Okazaki 444-8787, Aichi, Japan
- Graduate
Institute for Advanced Studies, SOKENDAI, Okazaki 444-8787, Aichi, Japan
| |
Collapse
|
8
|
Manning MC, Holcomb RE, Payne RW, Stillahn JM, Connolly BD, Katayama DS, Liu H, Matsuura JE, Murphy BM, Henry CS, Crommelin DJA. Stability of Protein Pharmaceuticals: Recent Advances. Pharm Res 2024; 41:1301-1367. [PMID: 38937372 DOI: 10.1007/s11095-024-03726-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024]
Abstract
There have been significant advances in the formulation and stabilization of proteins in the liquid state over the past years since our previous review. Our mechanistic understanding of protein-excipient interactions has increased, allowing one to develop formulations in a more rational fashion. The field has moved towards more complex and challenging formulations, such as high concentration formulations to allow for subcutaneous administration and co-formulation. While much of the published work has focused on mAbs, the principles appear to apply to any therapeutic protein, although mAbs clearly have some distinctive features. In this review, we first discuss chemical degradation reactions. This is followed by a section on physical instability issues. Then, more specific topics are addressed: instability induced by interactions with interfaces, predictive methods for physical stability and interplay between chemical and physical instability. The final parts are devoted to discussions how all the above impacts (co-)formulation strategies, in particular for high protein concentration solutions.'
Collapse
Affiliation(s)
- Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO, USA.
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA.
| | - Ryan E Holcomb
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Robert W Payne
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | | | | | | | | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | |
Collapse
|
9
|
Lapenna A, Dagallier C, Huille S, Tribet C. Poly(glutamic acid)-Based Viscosity Reducers for Concentrated Formulations of a Monoclonal IgG Antibody. Mol Pharm 2024; 21:982-991. [PMID: 38240032 PMCID: PMC10849046 DOI: 10.1021/acs.molpharmaceut.3c01159] [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: 12/08/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024]
Abstract
Above a concentration threshold, the viscosity of solutions of proteins increases abruptly, which hampers the injectability of therapeutic formulations. Concentrations above 200 g/L are an ideal goal for subcutaneous application of antibodies. Molecular additives, such as amino acids (e.g., arginine) help decrease the viscosity, but they are used at concentrations as high as about 200 mmol/L. We addressed the question of whether poly(amino acids) could be more efficient than small molecular additives. We observed marked fluidification of a model therapeutic monoclonal antibody (mAb) solution by poly(d,l-glutamic acid) and poly(l-glutamic acid) derivatives added at concentrations of <6.5 g/L (i.e., a mAb/polymer chain molar ratio between 4:1 and 1:1 mol/mol). The bare poly(glutamate) parent chains were compared with polyethylene glycol-grafted chains as PEGylation is a common way to enhance stability. Viscosity could be decreased to ∼20 mPa s as compared to values of ∼100 mPa s in the absence of polymers at 200 g/L mAb. Formation of complexes between the mAb and the polyglutamates was characterized by capillary electrophoresis analysis in dilute solutions (1 g/L mAb) and by observation of phase separation at higher concentrations, suggesting tight association at about 2:1 mol/mol mAb/polymer. Altogether, these results show that polyglutamate derivatives hold an untapped potential as an excipient for fluidification of concentrated protein solutions.
Collapse
Affiliation(s)
- Annamaria Lapenna
- Département
de Chimie, PASTEUR, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris 75005, France
| | - Camille Dagallier
- Biologics
Formulation & Process Development, Biologics
Drug Product Development Department, SANOFI R&D, 13 quai Jules Guesde- BP 14, Vitry-sur-Seine 94403, France
| | - Sylvain Huille
- Biologics
Formulation & Process Development, Biologics
Drug Product Development Department, SANOFI R&D, 13 quai Jules Guesde- BP 14, Vitry-sur-Seine 94403, France
| | - Christophe Tribet
- Département
de Chimie, PASTEUR, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris 75005, France
| |
Collapse
|
10
|
Gupta MN, Uversky VN. Biological importance of arginine: A comprehensive review of the roles in structure, disorder, and functionality of peptides and proteins. Int J Biol Macromol 2024; 257:128646. [PMID: 38061507 DOI: 10.1016/j.ijbiomac.2023.128646] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 01/26/2024]
Abstract
Arginine shows Jekyll and Hyde behavior in several respects. It participates in protein folding via ionic and H-bonds and cation-pi interactions; the charge and hydrophobicity of its side chain make it a disorder-promoting amino acid. Its methylation in histones; RNA binding proteins; chaperones regulates several cellular processes. The arginine-centric modifications are important in oncogenesis and as biomarkers in several cardiovascular diseases. The cross-links involving arginine in collagen and cornea are involved in pathogenesis of tissues but have also been useful in tissue engineering and wound-dressing materials. Arginine is a part of active site of several enzymes such as GTPases, peroxidases, and sulfotransferases. Its metabolic importance is obvious as it is involved in production of urea, NO, ornithine and citrulline. It can form unusual functional structures such as molecular tweezers in vitro and sprockets which engage DNA chains as part of histones in vivo. It has been used in design of cell-penetrating peptides as drugs. Arginine has been used as an excipient in both solid and injectable drug formulations; its role in suppressing opalescence due to liquid-liquid phase separation is particularly very promising. It has been known as a suppressor of protein aggregation during protein refolding. It has proved its usefulness in protein bioseparation processes like ion-exchange, hydrophobic and affinity chromatographies. Arginine is an amino acid, whose importance in biological sciences and biotechnology continues to grow in diverse ways.
Collapse
Affiliation(s)
- Munishwar Nath Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Hauz Khas, New Delhi 110016, India
| | - Vladimir N Uversky
- Department of Molecular Medicine, USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
11
|
Heisler J, Kovner D, Izadi S, Zarzar J, Carter PJ. Modulation of the high concentration viscosity of IgG 1 antibodies using clinically validated Fc mutations. MAbs 2024; 16:2379560. [PMID: 39028186 PMCID: PMC11262234 DOI: 10.1080/19420862.2024.2379560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/09/2024] [Indexed: 07/20/2024] Open
Abstract
The self-association of therapeutic antibodies can result in elevated viscosity and create problems in manufacturing and formulation, as well as limit delivery by subcutaneous injection. The high concentration viscosity of some antibodies has been reduced by variable domain mutations or by the addition of formulation excipients. In contrast, the impact of Fc mutations on antibody viscosity has been minimally explored. Here, we studied the effect of a panel of common and clinically validated Fc mutations on the viscosity of two closely related humanized IgG1, κ antibodies, omalizumab (anti-IgE) and trastuzumab (anti-HER2). Data presented here suggest that both Fab-Fab and Fab-Fc interactions contribute to the high viscosity of omalizumab, in a four-contact model of self-association. Most strikingly, the high viscosity of omalizumab (176 cP) was reduced 10.7- and 2.2-fold by Fc modifications for half-life extension (M252Y:S254T:T256E) and aglycosylation (N297G), respectively. Related single mutations (S254T and T256E) each reduced the viscosity of omalizumab by ~6-fold. An alternative half-life extension Fc mutant (M428L:N434S) had the opposite effect in increasing the viscosity of omalizumab by 1.5-fold. The low viscosity of trastuzumab (8.6 cP) was unchanged or increased by ≤ 2-fold by the different Fc variants. Molecular dynamics simulations provided mechanistic insight into the impact of Fc mutations in modulating electrostatic and hydrophobic surface properties as well as conformational stability of the Fc. This study demonstrates that high viscosity of some IgG1 antibodies can be mitigated by Fc mutations, and thereby offers an additional tool to help design future antibody therapeutics potentially suitable for subcutaneous delivery.
Collapse
Affiliation(s)
- Joel Heisler
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA, USA
| | - Daniel Kovner
- Department of Pharmaceutical Development, Genentech, Inc, South San Francisco, CA, USA
| | - Saeed Izadi
- Department of Pharmaceutical Development, Genentech, Inc, South San Francisco, CA, USA
| | - Jonathan Zarzar
- Department of Pharmaceutical Development, Genentech, Inc, South San Francisco, CA, USA
| | - Paul J. Carter
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
12
|
Vitharana S, Stillahn JM, Katayama DS, Henry CS, Manning MC. Application of Formulation Principles to Stability Issues Encountered During Processing, Manufacturing, and Storage of Drug Substance and Drug Product Protein Therapeutics. J Pharm Sci 2023; 112:2724-2751. [PMID: 37572779 DOI: 10.1016/j.xphs.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
The field of formulation and stabilization of protein therapeutics has become rather extensive. However, most of the focus has been on stabilization of the final drug product. Yet, proteins experience stress and degradation through the manufacturing process, starting with fermentaition. This review describes how formulation principles can be applied to stabilize biopharmaceutical proteins during bioprocessing and manufacturing, considering each unit operation involved in prepration of the drug substance. In addition, the impact of the container on stabilty is discussed as well.
Collapse
Affiliation(s)
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO 80534, USA; Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
13
|
Prass T, Garidel P, Blech M, Schäfer LV. Viscosity Prediction of High-Concentration Antibody Solutions with Atomistic Simulations. J Chem Inf Model 2023; 63:6129-6140. [PMID: 37757589 PMCID: PMC10565822 DOI: 10.1021/acs.jcim.3c00947] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Indexed: 09/29/2023]
Abstract
The computational prediction of the viscosity of dense protein solutions is highly desirable, for example, in the early development phase of high-concentration biopharmaceutical formulations where the material needed for experimental determination is typically limited. Here, we use large-scale atomistic molecular dynamics (MD) simulations with explicit solvation to de novo predict the dynamic viscosities of solutions of a monoclonal IgG1 antibody (mAb) from the pressure fluctuations using a Green-Kubo approach. The viscosities at simulated mAb concentrations of 200 and 250 mg/mL are compared to the experimental values, which we measured with rotational rheometry. The computational viscosity of 24 mPa·s at the mAb concentration of 250 mg/mL matches the experimental value of 23 mPa·s obtained at a concentration of 213 mg/mL, indicating slightly different effective concentrations (or activities) in the MD simulations and in the experiments. This difference is assigned to a slight underestimation of the effective mAb-mAb interactions in the simulations, leading to a too loose dynamic mAb network that governs the viscosity. Taken together, this study demonstrates the feasibility of all-atom MD simulations for predicting the properties of dense mAb solutions and provides detailed microscopic insights into the underlying molecular interactions. At the same time, it also shows that there is room for further improvements and highlights challenges, such as the massive sampling required for computing collective properties of dense biomolecular solutions in the high-viscosity regime with reasonable statistical precision.
Collapse
Affiliation(s)
- Tobias
M. Prass
- Center
for Theoretical Chemistry, Ruhr University
Bochum, D-44780 Bochum, Germany
| | - Patrick Garidel
- Boehringer
Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, D-88397 Biberach
an der Riss, Germany
| | - Michaela Blech
- Boehringer
Ingelheim Pharma GmbH & Co. KG, Innovation Unit, PDB, D-88397 Biberach
an der Riss, Germany
| | - Lars V. Schäfer
- Center
for Theoretical Chemistry, Ruhr University
Bochum, D-44780 Bochum, Germany
| |
Collapse
|
14
|
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
|
15
|
Kulakova A, Augustijn D, El Bialy I, Gentiluomo L, Greco ML, Hervø-Hansen S, Indrakumar S, Mahapatra S, Martinez Morales M, Pohl C, Polimeni M, Roche A, Svilenov HL, Tosstorff A, Zalar M, Curtis R, Derrick JP, Frieß W, Golovanov AP, Lund M, Nørgaard A, Khan TA, Peters GHJ, Pluen A, Roessner D, Streicher WW, van der Walle CF, Warwicker J, Uddin S, Winter G, Bukrinski JT, Rinnan Å, Harris P. Chemometrics in Protein Formulation: Stability Governed by Repulsion and Protein Unfolding. Mol Pharm 2023. [PMID: 37146162 DOI: 10.1021/acs.molpharmaceut.3c00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Therapeutic proteins can be challenging to develop due to their complexity and the requirement of an acceptable formulation to ensure patient safety and efficacy. To date, there is no universal formulation development strategy that can identify optimal formulation conditions for all types of proteins in a fast and reliable manner. In this work, high-throughput characterization, employing a toolbox of five techniques, was performed on 14 structurally different proteins formulated in 6 different buffer conditions and in the presence of 4 different excipients. Multivariate data analysis and chemometrics were used to analyze the data in an unbiased way. First, observed changes in stability were primarily determined by the individual protein. Second, pH and ionic strength are the two most important factors determining the physical stability of proteins, where there exists a significant statistical interaction between protein and pH/ionic strength. Additionally, we developed prediction methods by partial least-squares regression. Colloidal stability indicators are important for prediction of real-time stability, while conformational stability indicators are important for prediction of stability under accelerated stress conditions at 40 °C. In order to predict real-time storage stability, protein-protein repulsion and the initial monomer fraction are the most important properties to monitor.
Collapse
Affiliation(s)
- Alina Kulakova
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| | - Dillen Augustijn
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, Frederiksberg 1958, Denmark
| | - Inas El Bialy
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Lorenzo Gentiluomo
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
- Wyatt Technology Europe GmbH, Hochstrasse 18, Dernbach 56307, Germany
| | - Maria Laura Greco
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Stefan Hervø-Hansen
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund 22100, Sweden
| | - Sowmya Indrakumar
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| | | | - Marcello Martinez Morales
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Christin Pohl
- Novozymes A/S, Krogshoejvej 36, Bagsvaerd 2880, Denmark
| | - Marco Polimeni
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund 22100, Sweden
| | - Aisling Roche
- Department of Chemical Engineering, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Hristo L Svilenov
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Andreas Tosstorff
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Matja Zalar
- Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, and Manchester Institute of Biotechnology, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| | - Robin Curtis
- Department of Chemical Engineering, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Jeremy P Derrick
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, U.K
| | - Wolfgang Frieß
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Alexander P Golovanov
- Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, and Manchester Institute of Biotechnology, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| | - Mikael Lund
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund 22100, Sweden
| | | | - Tarik A Khan
- Pharmaceutical Development & Supplies, Pharma Technical Development Biologics Europe, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| | - Alain Pluen
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, U.K
| | - Dierk Roessner
- Wyatt Technology Europe GmbH, Hochstrasse 18, Dernbach 56307, Germany
| | | | - Christopher F van der Walle
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Jim Warwicker
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, U.K
| | - Shahid Uddin
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | | | - Åsmund Rinnan
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, Frederiksberg 1958, Denmark
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| |
Collapse
|
16
|
Heat treatment in the presence of arginine increases the emulsifying properties of soy proteins. Food Chem X 2023; 17:100567. [PMID: 36845474 PMCID: PMC9945471 DOI: 10.1016/j.fochx.2023.100567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/21/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
This study aimed to improve the emulsifying properties of commercial soy protein isolates (CSPIs). CSPIs were thermally denatured without additives (CSPI_H) and with arginine (CSPI_A), urea (CSPI_U), and guanidine hydrochloride (CSPI_G), which improve protein solubility to prevent aggregation. These additives were removed by dialysis, and the samples were lyophilized. CSPI_A resulted in high emulsifying properties. FT-IR analysis showed that the β-sheet content in CSPI_A was reduced compared to that of untreated CSPI (CSPI_F). Fluorescence analysis showed that the tryptophan-derived emission peak of CSPI_A shifted between CSPI_F and CSPI_H which was exposed to hydrophobic amino acid chains with aggregation. As a result, the structure of CSPI_A became moderately unfolded and exposed the hydrophobic amino acid chains without aggregation. The CSPI_A solution had a more reduced oil-water interface tension than other CSPIs. These results support that CSPI_A attaches efficiently to the oil-water interface and produces small, less flocculated emulsions.
Collapse
|
17
|
Ghosh I, Gutka H, Krause ME, Clemens R, Kashi RS. A systematic review of commercial high concentration antibody drug products approved in the US: formulation composition, dosage form design and primary packaging considerations. MAbs 2023; 15:2205540. [PMID: 37243580 DOI: 10.1080/19420862.2023.2205540] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/29/2023] Open
Abstract
Three critical aspects that define high concentration antibody products (HCAPs) are as follows: 1) formulation composition, 2) dosage form, and 3) primary packaging configuration. HCAPs have become successful in the therapeutic sector due to their unique advantage of allowing subcutaneous self-administration. Technical challenges, such as physical and chemical instability, viscosity, delivery volume limitations, and product immunogenicity, can hinder successful development and commercialization of HCAPs. Such challenges can be overcome by robust formulation and process development strategies, as well as rational selection of excipients and packaging components. We compiled and analyzed data from US Food and Drug Administration-approved and marketed HCAPs that are ≥100 mg/mL to identify trends in formulation composition and quality target product profile. This review presents our findings and discusses novel formulation and processing technologies that enable the development of improved HCAPs at ≥200 mg/mL. The observed trends can be used as a guide for further advancements in the development of HCAPs as more complex antibody-based modalities enter biologics product development.
Collapse
Affiliation(s)
- Indrajit Ghosh
- Sterile Product Development, Bristol Myers Squibb, New Brunswick, NJ, USA
| | - Hiten Gutka
- Sterile Product Development, Bristol Myers Squibb, New Brunswick, NJ, USA
| | - Mary E Krause
- Sterile Product Development, Bristol Myers Squibb, New Brunswick, NJ, USA
| | - Ryan Clemens
- College of Pharmacy, University of Illinois at Chicago, Chicago, USA
| | - Ramesh S Kashi
- Sterile Product Development, Bristol Myers Squibb, Summit, NJ, USA
| |
Collapse
|
18
|
Yang D, Walker LM. Synergistic Effects of Multiple Excipients on Controlling Viscosity of Concentrated Protein Dispersions. J Pharm Sci 2022; 112:1379-1387. [PMID: 36539064 DOI: 10.1016/j.xphs.2022.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Viscosity control is essential for the manufacturing and delivery of concentrated therapeutic proteins. Limited availability of the precious protein-based drugs hinders the characterization and screening of the formulation conditions with new types or different combinations of excipients. In this work, a droplet-based microfluidic device with incorporated multiple particle tracking microrheology (MPT) is developed to quantify the effects of two excipients, arginine hydrochloride (ArgHCl) and caffeine, on the viscosity of concentrated bovine gamma globulin (BGG) dispersions at two different values of pH. The effectiveness of both ArgHCl and caffeine show dependence on the BGG concentration and solution pH. The data set with high compositional resolution provides useful information to guide formulation with multiple viscosity-reducing excipients and quantification appropriate to start elucidating the connection to protein-protein interaction mechanisms. Overall, this work has demonstrated that the developed microfluidic approach has the potential to effectively assess the impact of multiple excipients on the viscosity and provide data for computational methods to predict viscosity for high concentration protein formulations.
Collapse
Affiliation(s)
- Deyu Yang
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, United States
| | - Lynn M Walker
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, United States.
| |
Collapse
|
19
|
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: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [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
|
20
|
Advanced Formulations/Drug Delivery Systems for Subcutaneous Delivery of Protein-Based Biotherapeutics. J Pharm Sci 2022; 111:2968-2982. [PMID: 36058255 DOI: 10.1016/j.xphs.2022.08.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 12/14/2022]
Abstract
Multiple advanced formulations and drug delivery systems (DDSs) have been developed to deliver protein-based biotherapeutics via the subcutaneous (SC) route. These formulations/DDSs include high-concentration solution, co-formulation of two or more proteins, large volume injection, protein cluster/complex, suspension, nanoparticle, microparticle, and hydrogel. These advanced systems provide clinical benefits related to efficacy and safety, but meanwhile, have more complicated formulations and manufacturing processes compared to conventional solution formulations. To develop a fit-for-purpose formulation/DDS for SC delivery, scientists need to consider multiple factors, such as the primary indication, targeted site, immunogenicity, compatibility, biopharmaceutics, patient compliance, etc. Next, they need to develop appropriate formulation (s) and manufacturing processes using the QbD principle and have a control strategy. This paper aims to provide a comprehensive review of advanced formulations/DDSs recently developed for SC delivery of proteins, as well as some knowledge gaps and potential strategies to narrow them through future research.
Collapse
|
21
|
Li S, Yoshizawa T, Shiramasa Y, Kanamaru M, Ide F, Kitamura K, Kashiwagi N, Sasahara N, Kitazawa S, Kitahara R. Mechanism underlying liquid-to-solid phase transition in fused in sarcoma liquid droplets. Phys Chem Chem Phys 2022; 24:19346-19353. [PMID: 35943083 DOI: 10.1039/d2cp02171d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The RNA-binding protein fused in sarcoma (FUS) forms ribonucleoprotein granules via liquid-liquid phase separation (LLPS) in the cytoplasm. The phase separation of FUS accelerates aberrant liquid-solid phase separation and leads to the onset of familial amyotrophic lateral sclerosis (ALS). We previously found that FUS forms two types of liquid condensates in equilibrium, specifically LP-LLPS (i.e., normal type) and HP-LLPS (i.e., aberrant type), each with different partial molar volumes. However, it is unclear how liquid condensates are converted to the pathogenic solid phase. Here, we report a mechanism underlying the aberrant liquid-to-solid phase transition of FUS liquid condensates and the inhibition of this transition with small molecules. We found that the liquid condensate formed via HP-LLPS had greatly reduced dynamics, which is a common feature of aged wild-type FUS droplets and the droplet-like assembly of the ALS patient-type FUS variant. The longer FUS remained on the HP-LLPS, the harder it was to transform it into a mixed state (i.e., one-phase). These results indicate that liquid-to-solid phase transition, namely the aging of droplets, is accelerated with HP-LLPS. Interestingly, arginine suppressed the aging of droplets and HP-LLPS formation more strongly than LP-LLPS formation. These data indicate that the formation of HP-LLPS via the one-phase state or LP-LLPS is a pathway leading to irreversible solid aggregates. Dopamine and pyrocatechol also suppressed HP-LLPS formation. Our data highlight the potential of HP-LLPS to be used as a therapeutic target and arginine as a plausible drug candidate for ALS-causing FUS.
Collapse
Affiliation(s)
- Shujie Li
- Graduate School of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Takuya Yoshizawa
- College of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Yutaro Shiramasa
- Graduate School of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Mako Kanamaru
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Fumika Ide
- Graduate School of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Keiji Kitamura
- Graduate School of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Norika Kashiwagi
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Naoya Sasahara
- Graduate School of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Soichiro Kitazawa
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Ryo Kitahara
- Graduate School of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan. .,College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| |
Collapse
|
22
|
Srivastava A, O'Dell C, Bolessa E, McLinden S, Fortin L, Deorkar N. Viscosity reduction and stability enhancement of monoclonal antibody formulations using derivatives of amino acids. J Pharm Sci 2022; 111:2848-2856. [DOI: 10.1016/j.xphs.2022.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 11/26/2022]
|
23
|
Blanco MA. Computational models for studying physical instabilities in high concentration biotherapeutic formulations. MAbs 2022; 14:2044744. [PMID: 35282775 PMCID: PMC8928847 DOI: 10.1080/19420862.2022.2044744] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Computational prediction of the behavior of concentrated protein solutions is particularly advantageous in early development stages of biotherapeutics when material availability is limited and a large set of formulation conditions needs to be explored. This review provides an overview of the different computational paradigms that have been successfully used in modeling undesirable physical behaviors of protein solutions with a particular emphasis on high-concentration drug formulations. This includes models ranging from all-atom simulations, coarse-grained representations to macro-scale mathematical descriptions used to study physical instability phenomena of protein solutions such as aggregation, elevated viscosity, and phase separation. These models are compared and summarized in the context of the physical processes and their underlying assumptions and limitations. A detailed analysis is also given for identifying protein interaction processes that are explicitly or implicitly considered in the different modeling approaches and particularly their relations to various formulation parameters. Lastly, many of the shortcomings of existing computational models are discussed, providing perspectives and possible directions toward an efficient computational framework for designing effective protein formulations.
Collapse
Affiliation(s)
- Marco A. Blanco
- Materials and Biophysical Characterization, Analytical R & D, Merck & Co., Inc, Kenilworth, NJ USA
| |
Collapse
|
24
|
Nakauchi Y, Nishinami S, Murakami Y, Ogura T, Kano H, Shiraki K. Opalescence Arising from Network Assembly in Antibody Solution. Mol Pharm 2022; 19:1160-1167. [PMID: 35274955 DOI: 10.1021/acs.molpharmaceut.1c00929] [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: 11/28/2022]
Abstract
Opalescence of therapeutic antibody solutions is one of the concerns in drug formulation. However, the mechanistic insights into the opalescence of antibody solutions remain unclear. Here, we investigated the assembly states of antibody molecules as a function of antibody concentration. The solutions of bovine gamma globulin and human immunoglobulin G at around 100 mg/mL showed the formation of submicron-scale network assemblies. The network assembly resulted in the appearance of opalescence with a transparent blue color without the precipitates of antibodies. Furthermore, the addition of trehalose and arginine, previously known to act as protein stabilizers and protein aggregation suppressors, was able to suppress the opalescence arising from the network assembly. These results will provide an important information for evaluating and improving protein formulations.
Collapse
Affiliation(s)
- Yoshitaka Nakauchi
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8573, Ibaraki, Japan
| | - Suguru Nishinami
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8573, Ibaraki, Japan
| | - Yusuke Murakami
- Ph.D. Program in Humanics, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Toshihiko Ogura
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Higashi, Tsukuba 305-8566, Ibaraki, Japan
| | - Hideaki Kano
- Department of Chemistry, Kyusyu University, 744, Motooka, Nishi-ku, Fukuoka-shi 819-0395, Fukuoka, Japan
| | - Kentaro Shiraki
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8573, Ibaraki, Japan
| |
Collapse
|
25
|
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
|
26
|
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: 34] [Impact Index Per Article: 8.5] [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
|
27
|
Droplet-Based Microfluidic Tool to Quantify Viscosity of Concentrating Protein Solutions. Pharm Res 2021; 38:1765-1775. [PMID: 34664208 DOI: 10.1007/s11095-021-03106-9] [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: 08/03/2021] [Accepted: 09/03/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Measurement of the viscosity of concentrated protein solutions is vital for the manufacture and delivery of protein therapeutics. Conventional methods for viscosity measurements require large solution volumes, creating a severe limitation during the early stage of protein development. The goal of this work is to develop a robust technique that requires minimal sample. METHODS In this work, a droplet-based microfluidic device is developed to quantify the viscosity of protein solutions while concentrating in micrometer-scale droplets. The technique requires only microliters of sample. The corresponding viscosity is characterized by multiple particle tracking microrheology (MPT). RESULTS We show that the viscosities quantified in the microfluidic device are consistent with macroscopic results measured by a conventional rheometer for poly(ethylene) glycol (PEG) solutions. The technique was further applied to quantify viscosities of well-studied lysozyme and bovine serum albumin (BSA) solutions. Comparison to both macroscopic measurements and models (Krieger-Dougherty model) demonstrate the validity of the approach. CONCLUSION The droplet-based microfluidic device provides accurate quantitative values of viscosity over a range of concentrations for protein solutions with small sample volumes (~ μL) and high compositional resolution. This device will be extended to study the effect of different excipients and other additives on the viscosity of protein solutions.
Collapse
|
28
|
Tsumura K, Hsu W, Mimura M, Horiuchi A, Shiraki K. Lowering the viscosity of a high-concentration antibody solution by protein-polyelectrolyte complex. J Biosci Bioeng 2021; 133:17-24. [PMID: 34629298 DOI: 10.1016/j.jbiosc.2021.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/21/2022]
Abstract
High-concentration and low-viscosity antibody formulations are necessary when administering these solutions subcutaneously (SC) due to limitations on injection volume. Here we show a method to decrease the viscosity of monoclonal antibody solution by protein-polyelectrolyte complex (PPC) with poly-l-glutamic acid (polyE). The viscosity of omalizumab solutions was 90 cP at the concentration of 150 mg/mL. In the presence of 20-50 mM polyE, the viscosity of PPC solution of 150 mg/mL omalizumab dramatically decreased below 10 cP due to the formation of crowded solution. The crowded state of PPC, named aggregated PPC (A-PPC), contained water droplets with a diameter of 10 μm or larger with low antibody concentrations. In the presence of 60 mM or more polyE, the omalizumab solution was transparent with the viscosity of 40 cP or less, named soluble PPC (S-PPC). More importantly, the solutions of both A-PPC and S-PPC were fully redissolved by the addition of phosphate saline buffer confirmed by secondary structure, the amount of aggregates, and binding activity to antigen.
Collapse
Affiliation(s)
- Keisuke Tsumura
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan
| | - WeiLin Hsu
- Research and Development Center, Terumo Corporation, Nakai-machi, Ashigarakami-gun, Kanagawa 259-0151, Japan
| | - Masahiro Mimura
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan
| | - Aiko Horiuchi
- Research and Development Center, Terumo Corporation, Nakai-machi, Ashigarakami-gun, Kanagawa 259-0151, Japan
| | - Kentaro Shiraki
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan.
| |
Collapse
|
29
|
Cloutier TK, Sudrik C, Mody N, Hasige SA, Trout BL. Molecular computations of preferential interactions of proline, arginine.HCl, and NaCl with IgG1 antibodies and their impact on aggregation and viscosity. MAbs 2021; 12:1816312. [PMID: 32938318 PMCID: PMC7531574 DOI: 10.1080/19420862.2020.1816312] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Preferential interactions of excipients with the antibody surface govern their effect on the stability of antibodies in solution. We probed the preferential interactions of proline, arginine.HCl (Arg.HCl), and NaCl with three therapeutically relevant IgG1 antibodies via experiment and simulation. With simulations, we examined how excipients interacted with different types of surface patches in the variable region (Fv). For example, proline interacted most strongly with aromatic surfaces, Arg.HCl was included near negative residues, and NaCl was excluded from negative residues and certain hydrophobic regions. The differences in interaction of different excipients with the same surface patch on an antibody may be responsible for variations in the antibody's aggregation, viscosity, and self-association behaviors in each excipient. Proline reduced self-association for all three antibodies and reduced aggregation for the antibody with an association-limited aggregation mechanism. The effects of Arg.HCl and NaCl on aggregation and viscosity were highly dependent on the surface charge distribution and the extent of exclusion from highly hydrophobic patches. At pH 5.5, both tended to increase the aggregation of an antibody with a strongly positive charge on the Fv, while only NaCl reduced the aggregation of the antibody with a large negative charge patch on the Fv. Arg.HCl reduced the viscosities of antibodies with either a hydrophobicity-driven mechanism or a charge-driven mechanism. Analysis of this data presents a framework for understanding how amino acid and ionic excipients interact with different protein surfaces, and how these interactions translate to the observed stability behavior.
Collapse
Affiliation(s)
- Theresa K Cloutier
- Department of Chemical Engineering, Massachusetts Institute of Technology , Cambridge, Maryland, USA
| | - Chaitanya Sudrik
- Department of Chemical Engineering, Massachusetts Institute of Technology , Cambridge, Maryland, USA
| | - Neil Mody
- Dosage Form Design and Development, AstraZeneca , Gaithersburg, Maryland, USA
| | - Sathish A Hasige
- Dosage Form Design and Development, AstraZeneca , Gaithersburg, Maryland, USA
| | - Bernhardt L Trout
- Department of Chemical Engineering, Massachusetts Institute of Technology , Cambridge, Maryland, USA
| |
Collapse
|
30
|
Bramham JE, Davies SA, Podmore A, Golovanov AP. Stability of a high-concentration monoclonal antibody solution produced by liquid-liquid phase separation. MAbs 2021; 13:1940666. [PMID: 34225583 PMCID: PMC8265796 DOI: 10.1080/19420862.2021.1940666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Subcutaneous injection of a low volume (<2 mL) high concentration (>100 mg/mL) formulation is an attractive administration strategy for monoclonal antibodies (mAbs) and other biopharmaceutical proteins. Using concentrated solutions may also be beneficial at various stages of bioprocessing. However, concentrating proteins by conventional techniques, such as ultrafiltration, can be time consuming and challenging. Isolation of the dense fraction produced by macroscopic liquid–liquid phase separation (LLPS) has been suggested as a means to produce high-concentration solutions, but practicality of this method, and the stability of the resulting protein solution have not previously been demonstrated. In this proof-of-concept study, we demonstrate that LLPS can be used to concentrate a mAb solution to >170 mg/mL. We show that the structure of the mAb is not altered by LLPS, and unperturbed mAb is recoverable following dilution of the dense fraction, as judged by 1H nuclear magnetic resonance spectroscopy. Finally, we show that the physical properties and stability of a model high concentration protein formulation obtained from the dense fraction can be improved, for example through the addition of the excipient arginine·glutamate. This results in a stable high-concentration protein formulation with reduced viscosity and no further macroscopic LLPS. Concentrating mAb solutions by LLPS represents a simple and effective technique to progress toward producing high-concentration protein formulations for bioprocessing or administration. Abbreviations Arginine·glutamate (Arg·Glu), Carr-Purcell-Meiboom-Gill (CPMG), critical temperature (TC), high-performance size-exclusion chromatography (HPSEC), liquid–liquid phase separation (LLPS), monoclonal antibody (mAb), nuclear magnetic resonance (NMR), transverse relaxation rate (R2)
Collapse
Affiliation(s)
- Jack E Bramham
- Manchester Institute of Biotechnology and Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Stephanie A Davies
- Dosage Form Design & Development, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Adrian Podmore
- Dosage Form Design & Development, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Alexander P Golovanov
- Manchester Institute of Biotechnology and Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| |
Collapse
|
31
|
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
|
32
|
Ye Y, Huo X, Yin Z. Protein-protein interactions at high concentrations: Effects of ArgHCl and NaCl on the stability, viscosity and aggregation mechanisms of protein solution. Int J Pharm 2021; 601:120535. [PMID: 33811966 DOI: 10.1016/j.ijpharm.2021.120535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/10/2021] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
The aim of this work was to use the diffusion coefficient ration (Dm/Dline) as a parameter to characterize the stability of protein at high concentration, to compare the effects of ArgHCl and NaCl on the interaction of highly concentrated proteins under different pH conditions, and to explore the correlation with protein stability. For this purpose, a high-concentration bovine serum albumin solution (BSA) was selected as the model system, and the diffusion coefficient, aggregation degree, conformational stability, and solution viscosity of the protein were studied by dynamic light scattering (DLS) and spectral detection techniques. The result showed that there was a significant correlation between the Dm/Dline and the protein aggregation. The Dm/Dline of the protein was minimum at pH 7.4, which corresponded to the maximum degree of aggregation and the highest solution viscosity. At pH 7.4, the hydrophobic interactions and the increased conformational stability of ArgHCl maximized the stability of the protein and reduced the viscosity of the solution by 69.3%. At pH 3.0, the strong charge shielding effect of ArgHCl and NaCl and the decreased conformational stability induced protein aggregation and the gel formation. These findings provided valuable insights into the mechanism of protein aggregation and the diffusion coefficient ration (Dm/Dline) could be a potential tool for the pre-formulation studies.
Collapse
Affiliation(s)
- Yalin Ye
- Key Laboratory of Drug Targeting and Novel Drug Delivery System Ministry of Education, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Xingli Huo
- Key Laboratory of Drug Targeting and Novel Drug Delivery System Ministry of Education, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Zongning Yin
- Key Laboratory of Drug Targeting and Novel Drug Delivery System Ministry of Education, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
33
|
Mieczkowski C, Cheng A, Fischmann T, Hsieh M, Baker J, Uchida M, Raghunathan G, Strickland C, Fayadat-Dilman L. Characterization and Modeling of Reversible Antibody Self-Association Provide Insights into Behavior, Prediction, and Correction. Antibodies (Basel) 2021; 10:antib10010008. [PMID: 33671864 PMCID: PMC7931086 DOI: 10.3390/antib10010008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/24/2020] [Accepted: 02/01/2021] [Indexed: 12/20/2022] Open
Abstract
Reversible antibody self-association, while having major developability and therapeutic implications, is not fully understood or readily predictable and correctable. For a strongly self-associating humanized mAb variant, resulting in unacceptable viscosity, the monovalent affinity of self-interaction was measured in the low μM range, typical of many specific and biologically relevant protein-protein interactions. A face-to-face interaction model extending across both the heavy-chain (HC) and light-chain (LC) Complementary Determining Regions (CDRs) was apparent from biochemical and mutagenesis approaches as well as computational modeling. Light scattering experiments involving individual mAb, Fc, Fab, and Fab'2 domains revealed that Fabs self-interact to form dimers, while bivalent mAb/Fab'2 forms lead to significant oligomerization. Site-directed mutagenesis of aromatic residues identified by homology model patch analysis and self-docking dramatically affected self-association, demonstrating the utility of these predictive approaches, while revealing a highly specific and tunable nature of self-binding modulated by single point mutations. Mutagenesis at these same key HC/LC CDR positions that affect self-interaction also typically abolished target binding with notable exceptions, clearly demonstrating the difficulties yet possibility of correcting self-association through engineering. Clear correlations were also observed between different methods used to assess self-interaction, such as Dynamic Light Scattering (DLS) and Affinity-Capture Self-Interaction Nanoparticle Spectroscopy (AC-SINS). Our findings advance our understanding of therapeutic protein and antibody self-association and offer insights into its prediction, evaluation and corrective mitigation to aid therapeutic development.
Collapse
Affiliation(s)
- Carl Mieczkowski
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., South San Francisco, CA 94080, USA; (C.M.); (M.H.); (J.B.); (M.U.); (G.R.); (L.F.-D.)
| | - Alan Cheng
- Discovery Chemistry, Modeling and Informatics, Merck & Co., Inc., South San Francisco, CA 94080, USA
- Correspondence: ; Tel.: +1-650-496-4834
| | - Thierry Fischmann
- Department of Chemistry, Modeling and Informatics, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (T.F.); (C.S.)
| | - Mark Hsieh
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., South San Francisco, CA 94080, USA; (C.M.); (M.H.); (J.B.); (M.U.); (G.R.); (L.F.-D.)
| | - Jeanne Baker
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., South San Francisco, CA 94080, USA; (C.M.); (M.H.); (J.B.); (M.U.); (G.R.); (L.F.-D.)
| | - Makiko Uchida
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., South San Francisco, CA 94080, USA; (C.M.); (M.H.); (J.B.); (M.U.); (G.R.); (L.F.-D.)
| | - Gopalan Raghunathan
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., South San Francisco, CA 94080, USA; (C.M.); (M.H.); (J.B.); (M.U.); (G.R.); (L.F.-D.)
| | - Corey Strickland
- Department of Chemistry, Modeling and Informatics, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (T.F.); (C.S.)
| | - Laurence Fayadat-Dilman
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., South San Francisco, CA 94080, USA; (C.M.); (M.H.); (J.B.); (M.U.); (G.R.); (L.F.-D.)
| |
Collapse
|
34
|
Chen SW, Tan D, Yang YS, Zhang W. Investigation of the effect of salt additives in Protein L affinity chromatography for the purification of tandem single-chain variable fragment bispecific antibodies. MAbs 2021; 12:1718440. [PMID: 31983280 PMCID: PMC6999846 DOI: 10.1080/19420862.2020.1718440] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Tandem single-chain variable fragment (scFv) bispecific antibodies (bsAb) are one of the most promising bsAb formats reported thus far. Yet, because of their increased aggregation propensity, high impurity content due to low expression level, smaller size and lack of the Fc region, it is challenging to isolate these products with high yield and purity within a limited number of purification steps in a scalable fashion. A robust purification process that is able to circumvent these issues is therefore of critical importance to allow effective isolation of this group of antibodies. We investigated the addition of sodium chloride (NaCl), calcium chloride (CaCl2), and L-arginine monohydrochloride (Arg·HCl) to the elution buffer of Protein L affinity chromatography, and propose here a novel mechanism for the modification of Protein L binding avidity that can lead to enhanced high molecular weight (HMW)-monomer separation, a preferential strengthening effect of the HMW-Protein L interaction compared to the monomer-Protein L interaction. In particular, we found Arg·HCl to be the most effective salt additive in terms of purity and recovery. The mechanism we propose is different from the widely reported chaotropic effect exerted by salt additives observed in Protein A chromatography. We also demonstrate here that a final eluate containing <1% HMW species and <100 ppm host cell proteins can be obtained within a two-step process with an overall yield of 65%, highlighting the promising suitability of Protein L affinity chromatography for the purification of kappa light chain-containing tandem scFv bsAb.
Collapse
Affiliation(s)
- Serene W Chen
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Darryl Tan
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Yuan Sheng Yang
- Animal Cell Technology Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Wei Zhang
- Downstream Processing Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| |
Collapse
|
35
|
Tilegenova C, Izadi S, Yin J, Huang CS, Wu J, Ellerman D, Hymowitz SG, Walters B, Salisbury C, Carter PJ. Dissecting the molecular basis of high viscosity of monospecific and bispecific IgG antibodies. MAbs 2021; 12:1692764. [PMID: 31779513 PMCID: PMC6927759 DOI: 10.1080/19420862.2019.1692764] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Some antibodies exhibit elevated viscosity at high concentrations, making them poorly suited for therapeutic applications requiring administration by injection such as subcutaneous or ocular delivery. Here we studied an anti-IL-13/IL-17 bispecific IgG4 antibody, which has anomalously high viscosity compared to its parent monospecific antibodies. The viscosity of the bispecific IgG4 in solution was decreased by only ~30% in the presence of NaCl, suggesting electrostatic interactions are insufficient to fully explain the drivers of viscosity. Intriguingly, addition of arginine-HCl reduced the viscosity of the bispecific IgG4 by ~50% to its parent IgG level. These data suggest that beyond electrostatics, additional types of interactions such as cation-π and/or π-π may contribute to high viscosity more significantly than previously understood. Molecular dynamics simulations of antibody fragments in the mixed solution of free arginine and explicit water were conducted to identify hotspots involved in self-interactions. Exposed surface aromatic amino acids displayed an increased number of contacts with arginine. Mutagenesis of the majority of aromatic residues pinpointed by molecular dynamics simulations effectively decreased the solution's viscosity when tested experimentally. This mutational method to reduce the viscosity of a bispecific antibody was extended to a monospecific anti-GCGR IgG1 antibody with elevated viscosity. In all cases, point mutants were readily identified that both reduced viscosity and retained antigen-binding affinity. These studies demonstrate a new approach to mitigate high viscosity of some antibodies by mutagenesis of surface-exposed aromatic residues on complementarity-determining regions that may facilitate some clinical applications.
Collapse
Affiliation(s)
| | - Saeed Izadi
- Early Stage Pharmaceutical Development, Genentech Inc., South San Francisco, CA, USA
| | - Jianping Yin
- Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | | | - Jiansheng Wu
- Protein Chemistry, Genentech Inc., South San Francisco, CA, USA
| | - Diego Ellerman
- Protein Chemistry, Genentech Inc., South San Francisco, CA, USA
| | - Sarah G Hymowitz
- Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Benjamin Walters
- Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Cleo Salisbury
- Early Stage Pharmaceutical Development, Genentech Inc., South San Francisco, CA, USA
| | - Paul J Carter
- Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| |
Collapse
|
36
|
Affiliation(s)
- James W. Swan
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge Massachusetts USA
| | - Samuel W. Winslow
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge Massachusetts USA
| | - William A. Tisdale
- Department of Chemical Engineering Massachusetts Institute of Technology Cambridge Massachusetts USA
| |
Collapse
|
37
|
Formulating monoclonal antibodies as powders for reconstitution at high concentration using spray-drying: Trehalose/amino acid combinations as reconstitution time reducing and stability improving formulations. Eur J Pharm Biopharm 2020; 156:131-142. [DOI: 10.1016/j.ejpb.2020.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/11/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
|
38
|
Sawant MS, Streu CN, Wu L, Tessier PM. Toward Drug-Like Multispecific Antibodies by Design. Int J Mol Sci 2020; 21:E7496. [PMID: 33053650 PMCID: PMC7589779 DOI: 10.3390/ijms21207496] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
The success of antibody therapeutics is strongly influenced by their multifunctional nature that couples antigen recognition mediated by their variable regions with effector functions and half-life extension mediated by a subset of their constant regions. Nevertheless, the monospecific IgG format is not optimal for many therapeutic applications, and this has led to the design of a vast number of unique multispecific antibody formats that enable targeting of multiple antigens or multiple epitopes on the same antigen. Despite the diversity of these formats, a common challenge in generating multispecific antibodies is that they display suboptimal physical and chemical properties relative to conventional IgGs and are more difficult to develop into therapeutics. Here we review advances in the design and engineering of multispecific antibodies with drug-like properties, including favorable stability, solubility, viscosity, specificity and pharmacokinetic properties. We also highlight emerging experimental and computational methods for improving the next generation of multispecific antibodies, as well as their constituent antibody fragments, with natural IgG-like properties. Finally, we identify several outstanding challenges that need to be addressed to increase the success of multispecific antibodies in the clinic.
Collapse
Affiliation(s)
- Manali S. Sawant
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (M.S.S.); (C.N.S.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Craig N. Streu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (M.S.S.); (C.N.S.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Chemistry, Albion College, Albion, MI 49224, USA
| | - Lina Wu
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter M. Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (M.S.S.); (C.N.S.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
39
|
Kulkarni SS, Patel SM, Bogner RH. Reconstitution Time for Highly Concentrated Lyophilized Proteins: Role of Formulation and Protein. J Pharm Sci 2020; 109:2975-2985. [DOI: 10.1016/j.xphs.2020.05.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/03/2020] [Accepted: 05/29/2020] [Indexed: 10/24/2022]
|
40
|
Resolving Liquid-Liquid Phase Separation for a Peptide Fused Monoclonal Antibody by Formulation Optimization. J Pharm Sci 2020; 110:738-745. [PMID: 32961238 DOI: 10.1016/j.xphs.2020.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/10/2020] [Accepted: 09/14/2020] [Indexed: 11/22/2022]
Abstract
Liquid-liquid phase separation (LLPS) of protein solutions has been usually related to strong protein-protein interactions (PPI) under certain conditions. For the first time, we observed the LLPS phenomenon for a novel protein modality, peptide-fused monoclonal antibody (pmAb). LLPS emerged within hours between pH 6.0 to 7.0 and disappeared when solution pH values decreased to pH 5.0 or lower. Negative values of interaction parameter (kD) and close to zero values of zeta potential (ζ) were correlated to LLPS appearance. However, between pH 6.0 to 7.0, a strong electrostatic repulsion force was expected to potentially avoid LLPS based on the sequence predicted pI value, 8.35. Surprisingly, this is significantly away from experimentally determined pI, 6.25, which readily attributes the LLPS appearances of pmAb to the attenuated electrostatic repulsion force. Such discrepancy between experiment and prediction reminds the necessity of actual measurement for a complicated modality like pmAb. Furthermore, significant protein degradation took place upon thermal stress at pH 5.0 or lower. Therefore, the effects of pH and selected excipients on the thermal stability of pmAb were further assessed. A formulation consisting of arginine at pH 6.5 successfully prevented the appearance of LLPS and enhanced its thermal stability at 40 °C for pmAb. In conclusion, we have reported LLPS for a pmAb and successfully resolved the issue by optimizing formulation with aids from PPI characterization.
Collapse
|
41
|
Izadi S, Patapoff TW, Walters BT. Multiscale Coarse-Grained Approach to Investigate Self-Association of Antibodies. Biophys J 2020; 118:2741-2754. [PMID: 32416079 DOI: 10.1016/j.bpj.2020.04.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/12/2020] [Accepted: 04/13/2020] [Indexed: 11/20/2022] Open
Abstract
Self-association of therapeutic monoclonal antibodies (mabs) are thought to modulate the undesirably high viscosity observed in their concentrated solutions. Computational prediction of such a self-association behavior is advantageous early during mab drug candidate selection when material availability is limited. Here, we present a coarse-grained (CG) simulation method that enables microsecond molecular dynamics simulations of full-length antibodies at high concentrations. The proposed approach differs from others in two ways: first, charges are assigned to CG beads in an effort to reproduce molecular multipole moments and charge asymmetry of full-length antibodies instead of only localized charges. This leads to great improvements in the agreement between CG and all-atom electrostatic fields. Second, the distinctive hydrophobic character of each antibody is incorporated through empirical adjustments to the short-range van der Waals terms dictated by cosolvent all-atom molecular dynamics simulations of antibody variable regions. CG simulations performed on a set of 15 different mabs reveal that diffusion coefficients in crowded environments are markedly impacted by intermolecular interactions. Diffusion coefficients computed from the simulations are in correlation with experimentally measured observables, including viscosities at a high concentration. Further, we show that the evaluation of electrostatic and hydrophobic characters of the mabs is useful in predicting the nonuniform effect of salt on the viscosity of mab solutions. This CG modeling approach is particularly applicable as a material-free screening tool for selecting antibody candidates with desirable viscosity properties.
Collapse
Affiliation(s)
- Saeed Izadi
- Pharmaceutical Development, Genentech, South San Francisco, California.
| | - Thomas W Patapoff
- Pharmaceutical Development, Genentech, South San Francisco, California
| | - Benjamin T Walters
- Biochemical and Cellular Pharmacology, Genentech, South San Francisco, California.
| |
Collapse
|
42
|
Orthogonal Techniques to Study the Effect of pH, Sucrose, and Arginine Salts on Monoclonal Antibody Physical Stability and Aggregation During Long-Term Storage. J Pharm Sci 2020; 109:584-594. [DOI: 10.1016/j.xphs.2019.10.065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/14/2019] [Accepted: 10/31/2019] [Indexed: 11/18/2022]
|
43
|
Chowdhury A, Guruprasad G, Chen AT, Karouta CA, Blanco MA, Truskett TM, Johnston KP. Protein-Protein Interactions, Clustering, and Rheology for Bovine IgG up to High Concentrations Characterized by Small Angle X-Ray Scattering and Molecular Dynamics Simulations. J Pharm Sci 2020; 109:696-708. [DOI: 10.1016/j.xphs.2019.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/18/2019] [Accepted: 11/01/2019] [Indexed: 01/23/2023]
|
44
|
Hu Y, Arora J, Joshi SB, Esfandiary R, Middaugh CR, Weis DD, Volkin DB. Characterization of Excipient Effects on Reversible Self-Association, Backbone Flexibility, and Solution Properties of an IgG1 Monoclonal Antibody at High Concentrations: Part 1. J Pharm Sci 2020; 109:340-352. [DOI: 10.1016/j.xphs.2019.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/13/2019] [Accepted: 06/04/2019] [Indexed: 12/21/2022]
|
45
|
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
|
46
|
Gong Y, Soleymani Abyaneh H, Drossis N, Niederquell A, Kuentz M, Leroux JC, de Haan HW, Gauthier MA. Ultra-sub-stoichiometric "Dynamic" Bioconjugation Reduces Viscosity by Disrupting Immunoglobulin Oligomerization. Biomacromolecules 2019; 20:3557-3565. [PMID: 31398010 DOI: 10.1021/acs.biomac.9b00867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Monoclonal antibodies (mAb) are a major focus of the pharmaceutical industry, and polyclonal immunoglobulin G (IgG) therapy is used to treat a wide variety of health conditions. As some individuals require mAb/IgG therapy their entire life, there is currently a great desire to formulate antibodies for bolus injection rather than infusion. However, to achieve the required doses, very concentrated antibody solutions may be required. Unfortunately, mAb/IgG self-assembly at high concentration can produce an unacceptably high viscosity for injection. To address this challenge, this study expands the concept of "dynamic covalent chemistry" to "dynamic bioconjugation" in order to reduce viscosity by interfering with antibody-antibody interactions. Ultra-sub-stoichiometric amounts of dynamic PEGylation agents (down to the nanomolar) significantly reduced the viscosity of concentrated antibody solutions by interfering with oligomerization.
Collapse
Affiliation(s)
- Yuhui Gong
- Swiss Federal Institute of Technology Zurich (ETHZ) , Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences , Vladimir-Prelog-Weg 3 , 8093 Zurich , Switzerland
| | - Hoda Soleymani Abyaneh
- Institut National de la Recherche Scientifique (INRS) , EMT Research Center , 1650 boul. Lionel-Boulet , Varennes , J3X 1S2 , Canada
| | - Nicole Drossis
- University of Ontario Institute of Technology , Faculty of Science , Oshawa , Ontario L1H 7K4 , Canada
| | - Andreas Niederquell
- University of Applied Sciences Northwestern Switzerland , School of Life Sciences, Institute of Pharma Technology , Hofackerstr. 30 , 4132 Muttenz , Switzerland
| | - Martin Kuentz
- University of Applied Sciences Northwestern Switzerland , School of Life Sciences, Institute of Pharma Technology , Hofackerstr. 30 , 4132 Muttenz , Switzerland
| | - Jean-Christophe Leroux
- Swiss Federal Institute of Technology Zurich (ETHZ) , Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences , Vladimir-Prelog-Weg 3 , 8093 Zurich , Switzerland
| | - Hendrick W de Haan
- University of Ontario Institute of Technology , Faculty of Science , Oshawa , Ontario L1H 7K4 , Canada
| | - Marc A Gauthier
- Institut National de la Recherche Scientifique (INRS) , EMT Research Center , 1650 boul. Lionel-Boulet , Varennes , J3X 1S2 , Canada
| |
Collapse
|
47
|
Xu AY, Castellanos MM, Mattison K, Krueger S, Curtis JE. Studying Excipient Modulated Physical Stability and Viscosity of Monoclonal Antibody Formulations Using Small-Angle Scattering. Mol Pharm 2019; 16:4319-4338. [DOI: 10.1021/acs.molpharmaceut.9b00687] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Amy Yuanyuan Xu
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, Maryland 20899, United States
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, Maryland 20850, United States
| | - Maria Monica Castellanos
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, Maryland 20899, United States
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, Maryland 20850, United States
| | - Kevin Mattison
- Malvern Panalytical, 117 Flanders Road, Westborough, Massachusetts 01581, United States
| | - Susan Krueger
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, Maryland 20899, United States
| | - Joseph E. Curtis
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
48
|
Hung JJ, Zeno WF, Chowdhury AA, Dear BJ, Ramachandran K, Nieto MP, Shay TY, Karouta CA, Hayden CC, Cheung JK, Truskett TM, Stachowiak JC, Johnston KP. Self-diffusion of a highly concentrated monoclonal antibody by fluorescence correlation spectroscopy: insight into protein-protein interactions and self-association. SOFT MATTER 2019; 15:6660-6676. [PMID: 31389467 DOI: 10.1039/c9sm01071h] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The dynamic behavior of monoclonal antibodies (mAbs) at high concentration provides insight into protein microstructure and protein-protein interactions (PPI) that influence solution viscosity and protein stability. At high concentration, interpretation of the collective-diffusion coefficient Dc, as determined by dynamic light scattering (DLS), is highly challenging given the complex hydrodynamics and PPI at close spacings. In contrast, self-diffusion of a tracer particle by Brownian motion is simpler to understand. Herein, we develop fluorescence correlation spectroscopy (FCS) for the measurement of the long-time self-diffusion of mAb2 over a wide range of concentrations and viscosities in multiple co-solute formulations with varying PPI. The normalized self-diffusion coefficient D0/Ds (equal to the microscopic relative viscosity ηeff/η0) was found to be smaller than η/η0. Smaller ratios of the microscopic to macroscopic viscosity (ηeff/η) are attributed to a combination of weaker PPI and less self-association. The interaction parameters extracted from fits of D0/Ds with a length scale dependent viscosity model agree with previous measurements of PPI by SLS and SAXS. Trends in the degree of self-association, estimated from ηeff/η with a microviscosity model, are consistent with oligomer sizes measured by SLS. Finally, measurements of collective diffusion and osmotic compressibility were combined with FCS data to demonstrate that the changes in self-diffusion between formulations are due primarily to changes in the protein-protein friction in these systems, and not to protein-solvent friction. Thus, FCS is a robust and accessible technique for measuring mAb self-diffusion, and, by extension, microviscosity, PPI and self-association that govern mAb solution dynamics.
Collapse
Affiliation(s)
- Jessica J Hung
- McKetta Department of Chemical Engineering, The University of Texas at Austin, 200 E Dean Keeton St Stop C0400, Austin, TX 78712, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Nishinami S, Kameda T, Arakawa T, Shiraki K. Hydantoin and Its Derivatives Reduce the Viscosity of Concentrated Antibody Formulations by Inhibiting Associations via Hydrophobic Amino Acid Residues. Ind Eng Chem Res 2019. [DOI: 10.1021/acs.iecr.9b01739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Suguru Nishinami
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan
| | - Tomoshi Kameda
- Artificial Intelligence Research Center, Advanced Industrial Science and Technology (AIST), 2-4-7 Aomi, Koto, Tokyo 135-0064, Japan
| | - Tsutomu Arakawa
- a Division of KBI Biopharma, Alliance Protein Laboratories, San Diego, California 92121, United States
| | - Kentaro Shiraki
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan
| |
Collapse
|
50
|
Enhancing Stability and Reducing Viscosity of a Monoclonal Antibody With Cosolutes by Weakening Protein-Protein Interactions. J Pharm Sci 2019; 108:2517-2526. [DOI: 10.1016/j.xphs.2019.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 02/12/2019] [Accepted: 03/01/2019] [Indexed: 12/22/2022]
|