1
|
Jacquot G, Lopez Navarro P, Grange C, Boudali L, Harlepp S, Pivot X, Detappe A. Landscape of Subcutaneous Administration Strategies for Monoclonal Antibodies in Oncology. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406604. [PMID: 39165046 DOI: 10.1002/adma.202406604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/15/2024] [Indexed: 08/22/2024]
Abstract
In recent decades, subcutaneous (SC) administration of monoclonal antibodies (mAbs) has emerged as a promising alternative to intravenous delivery in oncology, offering comparable therapeutic efficacy while addressing patient preferences. This perspective article provides an in-depth analysis of the technological landscape surrounding SC mAb administration in oncology. It outlines various technologies under evaluation across developmental stages, spanning from preclinical investigations to the integration of established methodologies in clinical practice. Additionally, this perspective article explores emerging trends and prospective trajectories, shedding light on the evolving landscape of SC mAb administration. Furthermore, it emphasizes key checkpoints related to quality attributes essential for optimizing mAb delivery via the SC route. This review serves as a valuable resource for researchers, clinicians, and healthcare policymakers, offering insights into the advancement of SC mAb administration in oncology and its implications for patient care.
Collapse
Affiliation(s)
- Guillaume Jacquot
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Pedro Lopez Navarro
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Coralie Grange
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Lotfi Boudali
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Sébastien Harlepp
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Alexandre Detappe
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
- Equipe de Synthèse Pour l'Analyse, Institut Pluridisciplinaire Hubert Curien (IPHC), UMR 7178 CNRS/University of Strasbourg, Strasbourg, Cedex 2, 67087, France
| |
Collapse
|
2
|
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
|
3
|
Mathias N, Huille S, Picci M, Mahoney RP, Pettis RJ, Case B, Helk B, Kang D, Shah R, Ma J, Bhattacharya D, Krishnamachari Y, Doucet D, Maksimovikj N, Babaee S, Garidel P, Esfandiary R, Gandhi R. Towards more tolerable subcutaneous administration: Review of contributing factors for improving combination product design. Adv Drug Deliv Rev 2024; 209:115301. [PMID: 38570141 DOI: 10.1016/j.addr.2024.115301] [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: 01/29/2024] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
Subcutaneous (SC) injections can be associated with local pain and discomfort that is subjective and may affect treatment adherence and overall patient experience. With innovations increasingly focused on finding ways to deliver higher doses and volumes (≥2 mL), there is a need to better understand the multiple intertwined factors that influence pain upon SC injection. As a priority for the SC Drug Development & Delivery Consortium, this manuscript provides a comprehensive review of known attributes from published literature that contribute to pain/discomfort upon SC injection from three perspectives: (1) device and delivery factors that cause physical pain, (2) formulation factors that trigger pain responses, and (3) human factors impacting pain perception. Leveraging the Consortium's collective expertise, we provide an assessment of the comparative and interdependent factors likely to impact SC injection pain. In addition, we offer expert insights and future perspectives to fill identified gaps in knowledge to help advance the development of patient-centric and well tolerated high-dose/high-volume SC drug delivery solutions.
Collapse
Affiliation(s)
- Neil Mathias
- Bristol-Myers Squibb, Co., 1 Squibb Dr, New Brunswick, NJ, 08901 USA
| | - Sylvain Huille
- Sanofi, 13 quai Jules Guesde, 94400 Vitry-Sur-Seine, France.
| | - Marie Picci
- Novartis Pharma AG, Fabrikstrasse 4, CH-4056 Basel, Switzerland
| | - Robert P Mahoney
- Comera Life Sciences, 12 Gill St, Suite 4650, Woburn, MA 01801 USA
| | - Ronald J Pettis
- Becton-Dickinson, 21 Davis Drive, Research Triangle Park, NC 27513 USA
| | - Brian Case
- KORU Medical Systems, 100 Corporate Dr, Mahwah, NJ 07430 USA
| | - Bernhard Helk
- Novartis Pharma AG, Werk Klybeck, WKL-681.4.42, CH-4057 Basel, Switzerland
| | - David Kang
- Halozyme Therapeutics, Inc., 12390 El Camino Real, San Diego, CA 92130 USA
| | - Ronak Shah
- Bristol-Myers Squibb, Co., 1 Squibb Dr, New Brunswick, NJ, 08901 USA
| | - Junchi Ma
- Johnson & Johnson Innovative Medicine, 200 Great Valley Pkwy, Malvern, PA 19355 USA
| | | | | | - Dany Doucet
- GSK, 1250 South Collegeville Road, Collegeville, PA 19426 USA
| | | | - Sahab Babaee
- Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, NJ 07065 USA
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397 Biberach/Riss, Germany
| | | | - Rajesh Gandhi
- Bristol-Myers Squibb, Co., 1 Squibb Dr, New Brunswick, NJ, 08901 USA
| |
Collapse
|
4
|
Rojekar S, Pallapati AR, Gimenez-Roig J, Korkmaz F, Sultana F, Sant D, Haeck C, Macdonald A, Kim SM, Rosen CJ, Barak O, Meseck M, Caminis J, Lizneva D, Yuen T, Zaidi M. Development and Biophysical Characterization of a Humanized FSH-Blocking Monoclonal Antibody Therapeutic Formulated at an Ultra-High Concentration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.11.540323. [PMID: 37214886 PMCID: PMC10197643 DOI: 10.1101/2023.05.11.540323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Highly concentrated antibody formulations are oftentimes required for subcutaneous, self-administered biologics. Here, we report the creation of a unique formulation for our first-in- class FSH-blocking humanized antibody, MS-Hu6, which we propose to move to the clinic for osteoporosis, obesity, and Alzheimer's disease. The studies were carried out using our Good Laboratory Practice (GLP) platform, compliant with the Code of Federal Regulations (Title 21, Part 58). We first used protein thermal shift, size exclusion chromatography, and dynamic light scattering to examine MS-Hu6 concentrations between 1 and 100 mg/mL. We found that thermal, monomeric, and colloidal stability of formulated MS-Hu6 was maintained at a concentration of 100 mg/mL. The addition of the antioxidant L-methionine and chelating agent disodium EDTA improved the formulation's long-term colloidal and thermal stability. Thermal stability was further confirmed by Nano differential scanning calorimetry (DSC). Physiochemical properties of formulated MS-Hu6, including viscosity, turbidity, and clarity, conformed with acceptable industry standards. That the structural integrity of MS-Hu6 in formulation was maintained was proven through Circular Dichroism (CD) and Fourier Transform Infrared (FTIR) spectroscopy. Three rapid freeze-thaw cycles at -80°C/25°C or -80°C/37°C further revealed excellent thermal and colloidal stability. Furthermore, formulated MS-Hu6, particularly its Fab domain, displayed thermal and monomeric storage stability for more than 90 days at 4°C and 25°C. Finally, the unfolding temperature (T m ) for formulated MS-Hu6 increased by >4.80°C upon binding to recombinant FSH, indicating highly specific ligand binding. Overall, we document the feasibility of developing a stable, manufacturable and transportable MS-Hu6 formulation at a ultra-high concentration at industry standards. The study should become a resource for developing biologic formulations in academic medical centers.
Collapse
|
5
|
Zhang C, Bye JW, Lui LH, Zhang H, Hales J, Brocchini S, Curtis RA, Dalby PA. Enhanced Thermal Stability and Reduced Aggregation in an Antibody Fab Fragment at Elevated Concentrations. Mol Pharm 2023; 20:2650-2661. [PMID: 37040431 PMCID: PMC10155210 DOI: 10.1021/acs.molpharmaceut.3c00081] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
The aggregation of protein therapeutics such as antibodies remains a major challenge in the biopharmaceutical industry. The present study aimed to characterize the impact of the protein concentration on the mechanisms and potential pathways for aggregation, using the antibody Fab fragment A33 as the model protein. Aggregation kinetics were determined for 0.05 to 100 mg/mL Fab A33, at 65 °C. A surprising trend was observed whereby increasing the concentration decreased the relative aggregation rate, ln(v) (% day-1), from 8.5 at 0.05 mg/mL to 4.4 at 100 mg/mL. The absolute aggregation rate (mol L-1 h-1) increased with the concentration following a rate order of approximately 1 up to a concentration of 25 mg/mL. Above this concentration, there was a transition to an apparently negative rate order of -1.1 up to 100 mg/mL. Several potential mechanisms were examined as possible explanations. A greater apparent conformational stability at 100 mg/mL was observed from an increase in the thermal transition midpoint (Tm) by 7-9 °C, relative to those at 1-4 mg/mL. The associated change in unfolding entropy (△Svh) also increased by 14-18% at 25-100 mg/mL, relative to those at 1-4 mg/mL, indicating reduced conformational flexibility in the native ensemble. Addition of Tween or the crowding agents Ficoll and dextran, showed that neither surface adsorption, diffusion limitations nor simple volume crowding affected the aggregation rate. Fitting of kinetic data to a wide range of mechanistic models implied a reversible two-state conformational switch mechanism from aggregation-prone monomers (N*) into non-aggregating native forms (N) at higher concentrations. kD measurements from DLS data also suggested a weak self-attraction while remaining colloidally stable, consistent with macromolecular self-crowding within weakly associated reversible oligomers. Such a model is also consistent with compaction of the native ensemble observed through changes in Tm and △Svh.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Biochemical Engineering, University College London, Gower Street, London WC1E 6BT, U.K
| | - Jordan W Bye
- School of Chemical Engineering and Analytical Science, The University of Manchester, Sackville Street, Manchester M13 9PL, U.K
| | - Lok H Lui
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, U.K
| | - Hongyu Zhang
- Department of Biochemical Engineering, University College London, Gower Street, London WC1E 6BT, U.K
| | - John Hales
- Department of Biochemical Engineering, University College London, Gower Street, London WC1E 6BT, U.K
| | - Steve Brocchini
- UCL School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, U.K
| | - Robin A Curtis
- School of Chemical Engineering and Analytical Science, The University of Manchester, Sackville Street, Manchester M13 9PL, U.K
| | - Paul A Dalby
- Department of Biochemical Engineering, University College London, Gower Street, London WC1E 6BT, U.K
| |
Collapse
|
6
|
Sant D, Rojekar S, Gera S, Pallapati AR, Gimenez-Roig J, Kuo TC, Padilla A, Korkmaz F, Cullen L, Chatterjee J, Shelly E, Meseck M, Miyashita S, Macdonald A, Sultana F, Barak O, Ryu V, Kim SM, Robinson C, Rosen CJ, Caminis J, Lizneva D, Haider S, Yuen T, Zaidi M. Optimizing a therapeutic humanized follicle-stimulating hormone-blocking antibody formulation by protein thermal shift assay. Ann N Y Acad Sci 2023; 1521:67-78. [PMID: 36628526 DOI: 10.1111/nyas.14952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Biopharmaceutical products are formulated using several Food and Drug Administration (FDA) approved excipients within the inactive ingredient limit to maintain their storage stability and shelf life. Here, we have screened and optimized different sets of excipient combinations to yield a thermally stable formulation for the humanized follicle-stimulating hormone (FSH)-blocking antibody, MS-Hu6. We used a protein thermal shift assay in which rising temperatures resulted in the maximal unfolding of the protein at the melting temperature (Tm ). To determine the buffer and pH for a stable solution, four different buffers with a pH range from 3 to 8 were screened. This resulted in maximal Tm s at pH 5.62 for Fab in phosphate buffer and at pH 6.85 for Fc in histidine buffer. Upon testing a range of salt concentrations, MS-Hu6 was found to be more stable at lower concentrations, likely due to reduced hydrophobic effects. Molecular dynamics simulations revealed a higher root-mean-square deviation with 1 mM than with 100 mM salt, indicating enhanced stability, as noted experimentally. Among the stabilizers tested, Tween 20 was found to yield the highest Tm and reversed the salt effect. Among several polyols/sugars, trehalose and sucrose were found to produce higher thermal stabilities. Finally, binding of recombinant human FSH to MS-Hu6 in a final formulation (20 mM phosphate buffer, 1 mM NaCl, 0.001% w/v Tween 20, and 260 mM trehalose) resulted in a thermal shift (increase in Tm ) for the Fab, but expectedly not in the Fc domain. Given that we used a low dose of MS-Hu6 (1 μM), the next challenge would be to determine whether 100-fold higher, industry-standard concentrations are equally stable.
Collapse
Affiliation(s)
- Damini Sant
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Satish Rojekar
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sakshi Gera
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anusha R Pallapati
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Judit Gimenez-Roig
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Tan-Chun Kuo
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ashley Padilla
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Liam Cullen
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jiya Chatterjee
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Eleanor Shelly
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marcia Meseck
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sari Miyashita
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anne Macdonald
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Farhath Sultana
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Orly Barak
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vitaly Ryu
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Se-Min Kim
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Cemre Robinson
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - John Caminis
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shozeb Haider
- Centre for Advanced Research Computing, School of Pharmacy, University College London, London, UK
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology and Departments of Medicine and of Pharmacology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
7
|
Larson NR, Wei Y, Cruz TA, Esfandiary R, Kalonia CK, Forrest ML, Middaugh CR. Conformational Changes and Drivers of Monoclonal Antibody Liquid-Liquid Phase Separation. J Pharm Sci 2023; 112:680-690. [PMID: 36306862 PMCID: PMC9974558 DOI: 10.1016/j.xphs.2022.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
Liquid-liquid phase separation is a phenomenon within biology whereby proteins can separate into dense and more dilute phases with distinct properties. Three antibodies that undergo liquid-liquid phase separation were characterized in the protein-rich and protein-poor phases. In comparison to the protein-poor phase, the protein-rich phase demonstrates more blue-shift tryptophan emissions and red-shifted amide I absorbances. Large changes involving conformational isomerization around disulfide bonds were observed using Raman spectroscopy. Amide I and protein fluorescence differences between the phases persisted to temperatures above the critical temperature but ceased at the temperature at which aggregation occurred. In addition, large changes occurred in the structural organization of water molecules within the protein-rich phase for all three antibodies. It is hypothesized that as the proteins have the same chemical potential in both phases, the protein viscosity is higher in the protein-rich phase resulting in slowed diffusion dependent protein aggregation in this phase. For all three antibodies we performed accelerated stability studies and found that the protein-rich phase aggregated at the same rate or slower than the protein-poor phase.
Collapse
Affiliation(s)
- Nicholas R Larson
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Ave, Lawrence, KS 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, 2030 Becker Driver, Lawrence, KS 66047
| | - Yangjie Wei
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Ave, Lawrence, KS 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, 2030 Becker Driver, Lawrence, KS 66047
| | - Thayana Araújo Cruz
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Ave, Lawrence, KS 66047; Federal University of Rio de Janeiro (UFRJ), IQ, Biochemistry Program, Ilha do Fundao, Rio de Janeiro, 21941-909, Brazil
| | - Reza Esfandiary
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Ave, Lawrence, KS 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, 2030 Becker Driver, Lawrence, KS 66047
| | - Cavan K Kalonia
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Ave, Lawrence, KS 66047; Macromolecule and Vaccine Stabilization Center, University of Kansas, 2030 Becker Driver, Lawrence, KS 66047
| | - M Laird Forrest
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Ave, Lawrence, KS 66047
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Ave, Lawrence, KS 66047.
| |
Collapse
|
8
|
Fuentes-Lemus E, Davies MJ. Effect of crowding, compartmentalization and nanodomains on protein modification and redox signaling - current state and future challenges. Free Radic Biol Med 2023; 196:81-92. [PMID: 36657730 DOI: 10.1016/j.freeradbiomed.2023.01.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/21/2022] [Accepted: 01/10/2023] [Indexed: 01/19/2023]
Abstract
Biological milieus are highly crowded and heterogeneous systems where organization of macromolecules within nanodomains (e.g. membraneless compartments) is vital to the regulation of metabolic processes. There is an increasing interest in understanding the effects that such packed environments have on different biochemical and biological processes. In this context, the redox biochemistry and redox signaling fields are moving towards investigating oxidative processes under conditions that exhibit these key features of biological systems in order to solve existing paradigms including those related to the generation and transmission of specific redox signals within and between cells in both normal physiology and under conditions of oxidative stress. This review outlines the effects that crowding, nanodomain formation and altered local viscosities can have on biochemical processes involving proteins, and then discusses some of the reactions and pathways involving proteins and oxidants that may, or are known to, be modulated by these factors. We postulate that knowledge of protein modification processes (e.g. kinetics, pathways and product formation) under conditions that mimic biological milieus, will provide a better understanding of the response of cells to endogenous and exogenous stressors, and their role in ageing, signaling, health and disease.
Collapse
Affiliation(s)
- Eduardo Fuentes-Lemus
- Department of Biomedical Sciences, Panum Institute, Blegdamsvej 3, University of Copenhagen, Copenhagen, 2200, Denmark.
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, Blegdamsvej 3, University of Copenhagen, Copenhagen, 2200, Denmark
| |
Collapse
|
9
|
Desai PG, Garidel P, Gbormittah FO, Kamen DE, Mills BJ, Narasimhan CN, Singh S, Stokes ESE, Walsh ER. An Intercompany Perspective on Practical Experiences of Predicting, Optimizing and Analyzing High Concentration Biologic Therapeutic Formulations. J Pharm Sci 2023; 112:359-369. [PMID: 36442683 DOI: 10.1016/j.xphs.2022.11.020] [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: 09/02/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022]
Abstract
Developing high-dose biologic drugs for subcutaneous injection often requires high-concentration formulations and optimizing viscosity, solubility, and stability while overcoming analytical, manufacturing, and administration challenges. To understand industry approaches for developing high-concentration formulations, the Formulation Workstream of the BioPhorum Development Group, an industry-wide consortium, conducted an inter-company collaborative exercise which included several surveys. This collaboration provided an industry perspective, experience, and insight into the practicalities for developing high-concentration biologics. To understand solubility and viscosity, companies desire predictive tools, but experience indicates that these are not reliable and experimental strategies are best. Similarly, most companies prefer accelerated and stress stability studies to in-silico or biophysical-based prediction methods to assess aggregation. In addition, optimization of primary container-closure and devices are pursued to mitigate challenges associated with high viscosity of the formulation. Formulation strategies including excipient selection and application of studies at low concentration to high-concentration formulations are reported. Finally, analytical approaches to high concentration formulations are presented. The survey suggests that although prediction of viscosity, solubility, and long-term stability is desirable, the outcome can be inconsistent and molecule dependent. Significant experimental studies are required to confirm robust product definition as modeling at low protein concentrations will not necessarily extrapolate to high concentration formulations.
Collapse
Affiliation(s)
- Preeti G Desai
- Bristol Myers Squibb, Sterile Product Development, 556 Morris Avenue, Summit, NJ 07901, USA
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH Co KG, Innovation Unit, PDB-TIP, 88397 Biberach an der Riss, Germany
| | - Francisca O Gbormittah
- GlaxoSmithKline, Strategic External Development, 1000 Winter Street North, Waltham, MA 02451, USA
| | - Douglas E Kamen
- Regeneron Pharmaceuticals Inc., Formulation Development, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Brittney J Mills
- AbbVie, NBE Drug Product Development, 1 N Waukegan Road, North Chicago, IL 60064, USA
| | | | - Shubhadra Singh
- GlaxoSmithKline R&D, Biopharmaceutical Product Sciences, Collegeville, PA 19426, USA
| | - Elaine S E Stokes
- BioPhorum, The Gridiron Building, 1 Pancras Square, London N1C 4AG UK.
| | - Erika R Walsh
- Merck & Co., Inc., Sterile and Specialty Products, Rahway, NJ, USA
| |
Collapse
|
10
|
Svilenov HL, Arosio P, Menzen T, Tessier P, Sormanni P. Approaches to expand the conventional toolbox for discovery and selection of antibodies with drug-like physicochemical properties. MAbs 2023; 15:2164459. [PMID: 36629855 PMCID: PMC9839375 DOI: 10.1080/19420862.2022.2164459] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/12/2023] Open
Abstract
Antibody drugs should exhibit not only high-binding affinity for their target antigens but also favorable physicochemical drug-like properties. Such drug-like biophysical properties are essential for the successful development of antibody drug products. The traditional approaches used in antibody drug development require significant experimentation to produce, optimize, and characterize many candidates. Therefore, it is attractive to integrate new methods that can optimize the process of selecting antibodies with both desired target-binding and drug-like biophysical properties. Here, we summarize a selection of techniques that can complement the conventional toolbox used to de-risk antibody drug development. These techniques can be integrated at different stages of the antibody development process to reduce the frequency of physicochemical liabilities in antibody libraries during initial discovery and to co-optimize multiple antibody features during early-stage antibody engineering and affinity maturation. Moreover, we highlight biophysical and computational approaches that can be used to predict physical degradation pathways relevant for long-term storage and in-use stability to reduce the need for extensive experimentation.
Collapse
Affiliation(s)
- Hristo L. Svilenov
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Gent, Belgium
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Tim Menzen
- Coriolis Pharma Research GmbH, Martinsried, 82152, Germany
| | - Peter Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Pietro Sormanni
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
11
|
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: 10] [Impact Index Per Article: 5.0] [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
|
12
|
Abstract
The aggregation propensity of monoclonal antibodies can be modified by adding different cosolutes into the solution. A simple coarse-grained model in the combination with the thermodynamic perturbation theory was used to predict cluster distribution and viscosity of the solutions of IgG4 monoclonal anibody in the presence of L-Arginine Hydrochloride. The data were analysed using binding polynomial to describe the binding of cosolute (Arginine) to the antibody molecule. The results show that by binding to the antibody molecule the cosolute occupies some of the binding sites of the antibody, and in this way reduces the amount of binding sites available to other antibody molecules. The aggregation propensity of the antibody molecules is therefore reduced.
Collapse
|
13
|
Goli VAR, Butreddy A. Biosimilar monoclonal antibodies: Challenges and approaches towards formulation. Chem Biol Interact 2022; 366:110116. [PMID: 36007632 DOI: 10.1016/j.cbi.2022.110116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/08/2022] [Accepted: 08/15/2022] [Indexed: 11/03/2022]
Abstract
Many biologic drug products, particularly monoclonal antibodies (mAbs), were off-patented between 2015 and 2020, and this process is continuing as the number of biologics approvals has increased. However, the availability of affordable biosimilars is delayed by secondary patents related to the formulation and manufacturing process. Therefore, an alternative formulation development is required to avoid infringement of formulation related patents. Several variables must be considered while developing alternative non-infringement formulations, including the time gap between the expiration of the molecule patent and the formulation patent, the ability not to infringe other secondary patents (process-related), and project timelines. As a part of life cycle management, innovator companies are adopting multiple strategies to delay biosimilar competition. Biosimilar companies could use the innovator formulation knowledge space to develop alternative formulations at the expense of time and cost. The present review discusses the key approaches in biosimilar formulation development, and further summarizes the use of innovator formulation knowledge space for biosimilar mAbs product development.
Collapse
Affiliation(s)
- Venkata Appa Reddy Goli
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S, Nagar, Punjab, 160062, India
| | - Arun Butreddy
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA.
| |
Collapse
|
14
|
Lauser KT, Rueter AL, Calabrese MA. Polysorbate identity and quantity dictate the extensional flow properties of protein‐excipient solutions. AIChE J 2022. [DOI: 10.1002/aic.17850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Kathleen T. Lauser
- Department of Chemical Engineering and Materials Science University of Minnesota Minneapolis Minnesota
| | - Amy L. Rueter
- Department of Chemical Engineering and Materials Science University of Minnesota Minneapolis Minnesota
| | - Michelle A. Calabrese
- Department of Chemical Engineering and Materials Science University of Minnesota Minneapolis Minnesota
| |
Collapse
|
15
|
Huang C, Chen L, Franzen L, Anderski J, Qian F. Spray-Dried Monoclonal Antibody Suspension for High-Concentration and Low-Viscosity Subcutaneous Injection. Mol Pharm 2022; 19:1505-1514. [PMID: 35417176 DOI: 10.1021/acs.molpharmaceut.2c00039] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Administration of highly concentrated monoclonal antibodies (mAbs) through injection is often not possible as the viscosity can be readily above 50 mPa·s when the concentration exceeds 150 mg/mL. Besides, highly concentrated mAb solutions always exhibit increased aggregation propensity and lower stability, which raise the difficulty for the successful development of highly concentrated mAb formulations. We hereby explored the possibility of suspension as another formulation form for high-concentration proteins to reduce viscosity and maintain stability. Specifically, we demonstrated that spray drying can serve as a process to prepare particles for suspension. Particles prepared from formulations with different mAb/trehalose mass ratios displayed good physical stability and antibody binding affinity, as indicated by circular dichroism, fluorescence spectroscopy, and surface plasmon resonance (SPR)-based bioassay analyses. During spray drying, a surface tension-dominated enrichment of mAb on the particle surface was observed, but this did not show a significant negative impact on mAb stability. Spray-dried particles were subsequently suspended into benzyl benzoate, and the resulting suspension showed good stability and a lower viscosity when compared to its counterpart solution. Furthermore, mAbs recovered from the suspension maintained their conformational structure. Our study demonstrated that the suspension displayed low viscosity and good physical stability, so it may offer novel opportunities for the preparation of highly concentrated protein formulations.
Collapse
Affiliation(s)
- Chengnan Huang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, P. R. China
| | - Linc Chen
- Bayer Healthcare Co. Ltd., Beijing, 100020, P. R. China
| | - Lutz Franzen
- Research & Development, Pharmaceuticals, Bayer AG, Wuppertal, 42096, Germany
| | - Juliane Anderski
- Research & Development, Pharmaceuticals, Bayer AG, Wuppertal, 42096, Germany
| | - Feng Qian
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
16
|
Effects of Monovalent Salt on Protein-Protein Interactions of Dilute and Concentrated Monoclonal Antibody Formulations. Antibodies (Basel) 2022; 11:antib11020024. [PMID: 35466277 PMCID: PMC9036246 DOI: 10.3390/antib11020024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
In this study, we used sodium chloride (NaCl) to extensively modulate non-specific protein-protein interactions (PPI) of a humanized anti-streptavidin monoclonal antibody class 2 molecule (ASA-IgG2). The changes in PPI with varying NaCl (CNaCl) and monoclonal antibody (mAb) concentration (CmAb) were assessed using the diffusion interaction parameter kD and second virial coefficient B22 measured from solutions with low to moderate CmAb. The effective structure factor S(q)eff measured from concentrated mAb solutions using small-angle X-ray and neutron scattering (SAXS/SANS) was also used to characterize the PPI. Our results found that the nature of net PPI changed not only with CNaCl, but also with increasing CmAb. As a result, parameters measured from dilute and concentrated mAb samples could lead to different predictions on the stability of mAb formulations. We also compared experimentally determined viscosity results with those predicted from interaction parameters, including kD and S(q)eff. The lack of a clear correlation between interaction parameters and measured viscosity values indicates that the relationship between viscosity and PPI is concentration-dependent. Collectively, the behavior of flexible mAb molecules in concentrated solutions may not be correctly predicted using models where proteins are considered to be uniform colloid particles defined by parameters derived from low CmAb.
Collapse
|
17
|
Lai PK, Gallegos A, Mody N, Sathish HA, Trout BL. Machine learning prediction of antibody aggregation and viscosity for high concentration formulation development of protein therapeutics. MAbs 2022; 14:2026208. [PMID: 35075980 PMCID: PMC8794240 DOI: 10.1080/19420862.2022.2026208] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Machine learning has been recently used to predict therapeutic antibody aggregation rates and viscosity at high concentrations (150 mg/ml). These works focused on commercially available antibodies, which may have been optimized for stability. In this study, we measured accelerated aggregation rates at 45°C and viscosity at 150 mg/ml for 20 preclinical and clinical-stage antibodies. Features obtained from molecular dynamics simulations of the full-length antibody and sequences were used for machine learning model construction. We found a k-nearest neighbors regression model with two features, spatial positive charge map on the CDRH2 and solvent-accessible surface area of hydrophobic residues on the variable fragment, gives the best performance for predicting antibody aggregation rates (r = 0.89). For the viscosity classification model, the model with the highest accuracy is a logistic regression model with two features, spatial negative charge map on the heavy chain variable region and spatial negative charge map on the light chain variable region. The accuracy and the area under precision recall curve of the classification model from validation tests are 0.86 and 0.70, respectively. In addition, we combined data from another 27 commercial mAbs to develop a viscosity predictive model. The best model is a logistic regression model with two features, number of hydrophobic residues on the light chain variable region and net charges on the light chain variable region. The accuracy and the area under precision recall curve of the classification model are 0.85 and 0.6, respectively. The aggregation rates and viscosity models can be used to predict antibody stability to facilitate pharmaceutical development.
Collapse
Affiliation(s)
- Pin-Kuang Lai
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, Hoboken, New Jersey, USA
| | - Austin Gallegos
- Dosage Form Design and Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Neil Mody
- Dosage Form Design and Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Hasige A Sathish
- Dosage Form Design and Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Bernhardt L Trout
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
18
|
Discovery of compounds with viscosity-reducing effects on biopharmaceutical formulations with monoclonal antibodies. Comput Struct Biotechnol J 2022; 20:5420-5429. [PMID: 36212536 PMCID: PMC9529560 DOI: 10.1016/j.csbj.2022.09.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022] Open
Abstract
Computational screening yielded 44 new viscosity-reducing agents on two model mAbs. Dual excipients for viscosity reduction and solution buffering were discovered. Compounds with three or more charges reduce the viscosity of model mAb formulations. Filtering based on physicochemical properties can be applied to other mAb formulations.
For the development of concentrated monoclonal antibody formulations for subcutaneous administration, the main challenge is the high viscosity of the solutions. To compensate for this, viscosity reducing agents are commonly used as excipients. Here, we applied two computational chemistry approaches to discover new viscosity-reducing agents: fingerprint similarity searching, and physicochemical property filtering. In total, 94 compounds were selected and experimentally evaluated on two model monoclonal antibodies, which led to the discovery of 44 new viscosity-reducing agents. Analysis of the results showed that using a simple filter that selects only compounds with three or more charge groups is a good ‘rule of thumb’ for selecting potential viscosity-reducing agents for two model monoclonal antibody formulations.
Collapse
|
19
|
Narhi LO, Chou DK, Christian TR, Gibson S, Jagannathan B, Jiskoot W, Jordan S, Sreedhara A, Waxman L, Das TK. Stress Factors in Primary Packaging, Transportation and Handling of Protein Drug Products and Their Impact on Product Quality. J Pharm Sci 2022; 111:887-902. [DOI: 10.1016/j.xphs.2022.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/15/2022] [Accepted: 01/16/2022] [Indexed: 12/15/2022]
|
20
|
Jiskoot W, Hawe A, Menzen T, Volkin DB, Crommelin DJA. Ongoing Challenges to Develop High Concentration Monoclonal Antibody-based Formulations for Subcutaneous Administration: Quo Vadis? J Pharm Sci 2021; 111:861-867. [PMID: 34813800 DOI: 10.1016/j.xphs.2021.11.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 11/26/2022]
Abstract
Although many subcutaneously (s.c.) delivered, high-concentration antibody formulations (HCAF) have received regulatory approval and are widely used commercially, formulation scientists are still presented with many ongoing challenges during HCAF development with new mAb and mAb-based candidates. Depending on the specific physicochemical and biological properties of a particular mAb-based molecule, such challenges vary from pharmaceutical attributes e.g., stability, viscosity, manufacturability, to clinical performance e.g., bioavailability, immunogenicity, and finally to patient experience e.g., preference for s.c. vs. intravenous delivery and/or preferred interactions with health-care professionals. This commentary focuses on one key formulation obstacle encountered during HCAF development: how to maximize the dose of the drug? We examine methodologies for increasing the protein concentration, increasing the volume delivered, or combining both approaches together. We discuss commonly encountered hurdles, i.e., physical protein instability and solution volume limitations, and we provide recommendations to formulation scientists to facilitate their development of s.c. administered HCAF with new mAb-based product candidates.
Collapse
Affiliation(s)
- W Jiskoot
- Coriolis Pharma Research GmbH, Fraunhoferstr. 18 b, 82152 Martinsried, Germany; Leiden Academic Center for Drug Research (LACDR), Leiden University, 2300 RA Leiden, the Netherlands
| | - Andrea Hawe
- Coriolis Pharma Research GmbH, Fraunhoferstr. 18 b, 82152 Martinsried, Germany
| | - Tim Menzen
- Coriolis Pharma Research GmbH, Fraunhoferstr. 18 b, 82152 Martinsried, Germany
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS 66047, USA
| | - Daan J A Crommelin
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands.
| |
Collapse
|
21
|
Hirschman J, Venkataramani D, Murphy MI, Patel SM, Du J, Amin S. Application of thin gap rheometry for high shear rate viscosity measurement in monoclonal antibody formulations. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Desai KG, Colandene JD, Gidh A, Hanslip S, Rastelli M. Syringe Filling of High-Concentration mAb Products Using Peristaltic Pump-Based Mechanism: Challenges and Mitigation Strategies. J Pharm Sci 2021; 111:562-576. [PMID: 34551352 DOI: 10.1016/j.xphs.2021.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 10/20/2022]
Abstract
Syringe filling of high-concentration mAb formulation during manufacturing of large-scale drug product batches may present challenges such as product deposition onto the area of the syringe barrel where the stopper is inserted, product splashing or dripping, droplets left after the fill cycle, filling needle clogging, product build-up inside the needle during line stoppages, variation in fill weight/volume, and potential impact on product quality attributes. In this article, a summary of these issues and approaches to overcome them are summarized. Potential failure modes of the syringe filling process and appropriate in-process controls are provided. In addition to developing the filling process or resolving manufacturing issues, the pharmaceutical company developing the product and associated drug product manufacturing process may want to implement long-term strategic approaches to support the portfolio progression. Potential long-term approaches such as use of a viscosity reducing formulation development approach, improving peristaltic filling technology performance, building small-scale filling capability and establishing a streamlined filling process management cycle are also summarized. The aspects summarized in this article may be used to develop a robust filling process and control strategy for high-concentration mAb products and implement long-term strategic approaches to support the portfolio progression.
Collapse
Affiliation(s)
- Kashappa Goud Desai
- Biopharmaceutical Product Sciences and Device Engineering, GlaxoSmithKline, 1250 S Collegeville Road, Collegeville, PA 19425, USA.
| | - James D Colandene
- Biopharmaceutical Product Sciences and Device Engineering, GlaxoSmithKline, 1250 S Collegeville Road, Collegeville, PA 19425, USA
| | - Aarti Gidh
- Biopharmaceutical Product Sciences and Device Engineering, GlaxoSmithKline, 1250 S Collegeville Road, Collegeville, PA 19425, USA
| | - Simon Hanslip
- Manufacturing Science and Technology, Specialty Large Molecule, GlaxoSmithKline, Harmire Road, Barnard Castle DL12 8DT, UK
| | - Massimo Rastelli
- New Product Introduction and Product Technology, Pharma Supply Chain, GlaxoSmithKline, Strada Asolana 90, San Polo di Torrile, Parma 43046, Italy
| |
Collapse
|
23
|
Eaton M, Que Z, Zhang J, Beck K, Shi R, McDermott J, Ladisch M, Yang Y. Multi-Electrode Array of Sensory Neurons as an In Vitro Platform to Identify the Nociceptive Response to Pharmaceutical Buffer Systems of Injectable Biologics. Pharm Res 2021; 38:1179-1186. [PMID: 34244893 DOI: 10.1007/s11095-021-03075-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/14/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Pharmaceutical buffer systems, especially for injectable biologics such as monoclonal antibodies, are an important component of successful FDA-approved medications. Clinical studies indicate that buffer components may be contributing factors for increased injection site pain. METHODS To determine the potential nociceptive effects of clinically relevant buffer systems, we developed an in vitro multi-electrode array (MEA) based recording system of rodent dorsal root ganglia (DRG) sensory neuron cell culture. This system monitors sensory neuron activity/firing as a surrogate of nociception when challenged with buffer components used in formulating monoclonal antibodies and other injectable biologics. RESULTS We show that citrate salt and citrate mannitol buffer systems cause an increase in mean firing rate, burst frequency, and burst duration in DRG sensory neurons, unlike histidine or saline buffer systems at the same pH value. Lowering the concentration of citrate leads to a lower firing intensity of DRG sensory neurons. CONCLUSION Increased activity/firing of DRG sensory neurons has been suggested as a key feature underlying nociception. Our results support the utility of an in vitro MEA assay with cultured DRG sensory neurons to probe the nociceptive potential of clinically relevant buffer components used in injectable biologics.
Collapse
Affiliation(s)
- Muriel Eaton
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, Hall for Discovery and Learning Research (DLR), 207 S Martin Jischke Dr., West Lafayette, Indiana, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Zhefu Que
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, Hall for Discovery and Learning Research (DLR), 207 S Martin Jischke Dr., West Lafayette, Indiana, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Jingliang Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, Hall for Discovery and Learning Research (DLR), 207 S Martin Jischke Dr., West Lafayette, Indiana, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA
| | - Kaethe Beck
- Office of the Executive Vice President for Research and Partnerships, Purdue University, West Lafayette, Indiana, USA
| | - Riyi Shi
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA.,Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA
| | - Jeff McDermott
- Eli Lilly and Company, Delivery Devices and Connected Solutions, Indianapolis, Indiana, USA
| | - Michael Ladisch
- Department of Agricultural and Biological Engineering, College of Engineering; Weldon School of Biomedical Engineering; and Laboratory of Renewable Resources Engineering (LORRE), Purdue University, 225 S. University Street, West Lafayette, Indiana, USA.
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, Hall for Discovery and Learning Research (DLR), 207 S Martin Jischke Dr., West Lafayette, Indiana, USA. .,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
24
|
Wu HH, Garidel P, Michaela B. HP-β-CD for the formulation of IgG and Ig-based biotherapeutics. Int J Pharm 2021; 601:120531. [PMID: 33775727 DOI: 10.1016/j.ijpharm.2021.120531] [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: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/20/2021] [Indexed: 12/14/2022]
Abstract
The main challenge to develop HCF for IgG and Ig-based therapeutics is to achieve essential solubility, viscosity and stability of these molecules in order to maintain product quality and meet regulatory requirement during manufacturing, production, storage, shipment and administration processes. The commonly used and FDA approved excipients for IgG and Ig -based therapeutics may no longer fulfil the challenge of HCF development for these molecules to certain extent, especially for some complex Ig-based platforms. 2-Hydroxypropyl beta-cyclodextrin (HP-β-CD) is one of the promising excipients applied recently for HCF development of IgG and Ig-based therapeutics although it has been used for formulation of small synthesized chemical drugs for more than thirty years. This review describes essential aspects about application of HP-β-CD as excipient in pharmaceutical formulation, including physico-chemical properties of HP-β-CD, supply chain, regulatory, patent landscape, marketed drugs with HP-β-CD, analytics and analytical challenges, stability and control strategies, and safety concerns. It also provides an overview of different studies, and outcomes thereof, regarding formulation development for IgGs and Ig-based molecules in liquid and solid (lyophilized) dosage forms with HP-β-CD. The review specifically highlights the challenges for formulation manufacturing of IgG and Ig-based therapeutics with HP-β-CD and identifies areas for future work in pharmaceutical and formulation development.
Collapse
Affiliation(s)
- Helen Haixia Wu
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, Pharmaceutical Development Biologicals, Biberach an der Riss, Germany.
| | - Patrick Garidel
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, Pharmaceutical Development Biologicals, Biberach an der Riss, Germany
| | - Blech Michaela
- Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, Pharmaceutical Development Biologicals, Biberach an der Riss, Germany
| |
Collapse
|
25
|
Protein intrinsic viscosity determination with the Viscosizer TD instrument: reaching beyond the initially expected applications. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2021; 50:587-595. [PMID: 33486532 DOI: 10.1007/s00249-020-01492-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/03/2020] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
Intrinsic viscosity is a key hydrodynamic parameter to understand molecular structure and hydration, as well as intramolecular interactions. Commercially available instruments measure intrinsic viscosity by recording the macromolecular mobility in a capillary. These instruments monitor Taylor dispersion using an absorbance or fluorescence detector. By design, these instruments behave like U-tube viscometers. To our knowledge, there are no studies to date showing that the Viscosizer TD instrument (Malvern-Panalytical) is able to measure the intrinsic viscosity of macromolecules. In this study, we then performed our assays on the Poly(ethylene oxide) polymer (PEO), used classically as a standard for viscometry measurements and on three model proteins: the bovine serum albumin (BSA), the bevacizumab monoclonal antibody, and the RTX Repeat Domain (RD) of the adenylate cyclase toxin of Bordetella pertussis (CyaA). The presence of P20 in the samples is critical to get reliable results. The data obtained with our in-house protocol show a strong correlation with intrinsic viscosity values obtained using conventional techniques. However, with respect to them, our measurements could be performed at relatively low concentrations, between 2 and 5 mg/ml, using only 7 µL per injection. Altogether, our results show that the Viscosizer TD instrument is able to measure intrinsic viscosities in a straightforward manner. This simple and innovative approach should give a new boost to intrinsic viscosity measurements and should reignite the interest of biophysicists, immunologists, structural biologists and other researchers for this key physicochemical parameter.
Collapse
|
26
|
Rodrigues D, Tanenbaum LM, Thirumangalathu R, Somani S, Zhang K, Kumar V, Amin K, Thakkar SV. Product-Specific Impact of Viscosity Modulating Formulation Excipients During Ultra-High Concentration Biotherapeutics Drug Product Development. J Pharm Sci 2020; 110:1077-1082. [PMID: 33340533 DOI: 10.1016/j.xphs.2020.12.016] [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: 10/06/2020] [Revised: 11/16/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022]
Abstract
Developing ultra-high concentration biotherapeutics drug products can be challenging due to increased viscosity, processing, and stability issues. Excipients used to alleviate these concerns are traditionally evaluated at lower protein concentrations. This study investigates whether classically known modulators of stability and viscosity at low (<50 mg/mL) to high (>50 - 150 mg/mL) protein concentrations are beneficial in ultra-high (>150 mg/mL) concentration protein formulations and drug products. This study evaluates the effect of arginine monohydrochloride, proline, and lysine monohydrochloride on viscosity and concentratability at different high and ultra-high protein concentrations using a monoclonal antibody, mAbN, formulation as a candidate protein system. The effect of excipients on the viscosity and concentratability (rate and extent) was different at high versus ultra-high protein concentrations. These results highlight that classical excipients in literature known to modulate protein interactions at low protein concentrations and reduce viscosity at high protein concentrations may need to be evaluated at target protein concentrations in a product-specific manner while developing ultra-high concentration biologics drug products.
Collapse
Affiliation(s)
- Danika Rodrigues
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, Pennsylvania 19355
| | - Laura M Tanenbaum
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, Pennsylvania 19355
| | - Renuka Thirumangalathu
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, Pennsylvania 19355
| | - Sandeep Somani
- Discovery Sciences, Janssen Research and Development (Janssen R&D), Spring House, Pennsylvania 19477
| | - Kai Zhang
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, Pennsylvania 19355
| | - Vineet Kumar
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, Pennsylvania 19355
| | - Ketan Amin
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, Pennsylvania 19355
| | - Santosh V Thakkar
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, Pennsylvania 19355; BioTherapeutics Cell and Developability Sciences (BioTD CDS), Janssen Research and Development (Janssen R&D), Spring House, Pennsylvania 19477.
| |
Collapse
|
27
|
Carrara SC, Ulitzka M, Grzeschik J, Kornmann H, Hock B, Kolmar H. From cell line development to the formulated drug product: The art of manufacturing therapeutic monoclonal antibodies. Int J Pharm 2020; 594:120164. [PMID: 33309833 DOI: 10.1016/j.ijpharm.2020.120164] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/23/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Therapeutic monoclonal antibodies and related products have steadily grown to become the dominant product class within the biopharmaceutical market. Production of antibodies requires special precautions to ensure safety and efficacy of the product. In particular, minimizing antibody product heterogeneity is crucial as drug substance variants may impair the activity, efficacy, safety, and pharmacokinetic properties of an antibody, consequently resulting in the failure of a product in pre-clinical and clinical development. This review will cover the manufacturing and formulation challenges and advances of therapeutic monoclonal antibodies, focusing on improved processes to minimize variants and ensure batch-to-batch consistency. Processes put in place by regulatory agencies, such as Quality-by-Design (QbD) and current Good Manufacturing Practices (cGMP), and how their implementation has aided drug development in pharmaceutical companies will be reviewed. Advances in formulation and considerations on the intended use of a therapeutic antibody, including the route of administration and patient compliance, will be discussed.
Collapse
Affiliation(s)
- Stefania C Carrara
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Ferring Darmstadt Laboratory, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Michael Ulitzka
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany; Ferring Darmstadt Laboratory, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Darmstadt Laboratory, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
| | - Henri Kornmann
- Ferring International Center SA, CH-1162 Saint-Prex, Switzerland
| | - Björn Hock
- Ferring International Center SA, CH-1162 Saint-Prex, Switzerland.
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany.
| |
Collapse
|
28
|
Kamerzell TJ, Middaugh CR. Prediction Machines: Applied Machine Learning for Therapeutic Protein Design and Development. J Pharm Sci 2020; 110:665-681. [PMID: 33278409 DOI: 10.1016/j.xphs.2020.11.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/27/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
The rapid growth in technological advances and quantity of scientific data over the past decade has led to several challenges including data storage and analysis. Accurate models of complex datasets were previously difficult to develop and interpret. However, improvements in machine learning algorithms have since enabled unparalleled classification and prediction capabilities. The application of machine learning can be seen throughout diverse industries due to their ease of use and interpretability. In this review, we describe popular machine learning algorithms and highlight their application in pharmaceutical protein development. Machine learning models have now been applied to better understand the nonlinear concentration dependent viscosity of protein solutions, predict protein oxidation and deamidation rates, classify sub-visible particles and compare the physical stability of proteins. We also applied several machine learning algorithms using previously published data and describe models with improved predictions and classification. The authors hope that this review can be used as a resource to others and encourage continued application of machine learning algorithms to problems in pharmaceutical protein development.
Collapse
Affiliation(s)
- Tim J Kamerzell
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA; Division of Internal Medicine, HCA MidWest Health, Overland Park, KS, USA.
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA
| |
Collapse
|
29
|
Kollár É, Balázs B, Tari T, Siró I. Development challenges of high concentration monoclonal antibody formulations. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 37:31-40. [PMID: 34895653 DOI: 10.1016/j.ddtec.2020.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/17/2020] [Accepted: 08/31/2020] [Indexed: 01/09/2023]
Abstract
High concentration monoclonal antibody drug products represent a special segment of biopharmaceuticals. In contrast to other monoclonal antibody products, high concentration monoclonal antibodies are injected subcutaneously helping increase patient compliance and reduce the number of hospital patient visits. It is important to note that a high protein concentration (≥50 mg/mL) poses a challenge from a product development perspective. Colloidal properties, physical and chemical protein stability should be considered during formulation, primary packaging and manufacturing process development as well as optimization of other dosage form-related parameters. The aim of such development work is to obtain a drug product capable of maintaining appropriate protein structure throughout its shelf-life and ensure proper and accurate dosage upon administration.
Collapse
Affiliation(s)
- Éva Kollár
- Department of Biotechnology Development, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary.
| | - Boglárka Balázs
- Department of Biotechnology Development, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
| | - Tímea Tari
- Department of Biotechnology Development, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
| | - István Siró
- Department of Biotechnology Development, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
| |
Collapse
|
30
|
Somani S, Jo S, Thirumangalathu R, Rodrigues D, Tanenbaum LM, Amin K, MacKerell AD, Thakkar SV. Toward Biotherapeutics Formulation Composition Engineering using Site-Identification by Ligand Competitive Saturation (SILCS). J Pharm Sci 2020; 110:1103-1110. [PMID: 33137372 DOI: 10.1016/j.xphs.2020.10.051] [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: 07/16/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 10/23/2022]
Abstract
Formulation of protein-based therapeutics employ advanced formulation and analytical technologies for screening various parameters such as buffer, pH, and excipients. At a molecular level, physico-chemical properties of a protein formulation depend on self-interaction between protein molecules, protein-solvent and protein-excipient interactions. This work describes a novel in silico approach, SILCS-Biologics, for structure-based modeling of protein formulations. SILCS Biologics is based on the Site-Identification by Ligand Competitive Saturation (SILCS) technology and enables modeling of interactions among different components of a formulation at an atomistic level while accounting for protein flexibility. It predicts potential hotspot regions on the protein surface for protein-protein and protein-excipient interactions. Here we apply SILCS-Biologics on a Fab domain of a monoclonal antibody (mAbN) to model Fab-Fab interactions and interactions with three amino acid excipients, namely, arginine HCl, proline and lysine HCl. Experiments on 100 mg/ml formulations of mAbN showed that arginine increased, lysine reduced, and proline did not impact viscosity. We use SILCS-Biologics modeling to explore a structure-based hypothesis for the viscosity modulating effect of these excipients. Current efforts are aimed at further validation of this novel computational framework and expanding the scope to model full mAb and other protein therapeutics.
Collapse
Affiliation(s)
- Sandeep Somani
- Discovery Sciences, Janssen Research and Development (Janssen R&D), Spring House, PA 19477, USA
| | | | - Renuka Thirumangalathu
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, PA 19355, USA
| | - Danika Rodrigues
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, PA 19355, USA
| | - Laura M Tanenbaum
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, PA 19355, USA
| | - Ketan Amin
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, PA 19355, USA
| | - Alexander D MacKerell
- SilcsBio LLC, Baltimore, MD 21202, USA; Computer-Aided Drug Design Center, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA.
| | - Santosh V Thakkar
- BioTherapeutics Drug Product Development (BioTD DPD), Janssen Research and Development (Janssen R&D), Malvern, PA 19355, USA; BioTherapeutics Cell and Developability Sciences (BioTD CDS), Janssen Research and Development (Janssen R&D), Spring House, PA 19477, USA.
| |
Collapse
|
31
|
Ren CD, Qi W, Wyatt EA, Yeary J, Westland K, Berke M, Rathore N. Application of a High Throughput and Automated Workflow to Therapeutic Protein Formulation Development. J Pharm Sci 2020; 110:1130-1141. [PMID: 33203511 DOI: 10.1016/j.xphs.2020.10.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/25/2020] [Accepted: 10/19/2020] [Indexed: 10/23/2022]
Abstract
Rapid and efficient formulation development is critical to successfully bringing therapeutic protein drug products into a competitive market under increasingly aggressive timelines. Conventional application of high throughput techniques for formulation development have been limited to lower protein concentrations, which are not applicable to late stage development of high concentration therapeutics. In this work, we present a high throughput (HT) formulation workflow that enables screening at representative concentrations via integration of a micro-buffer exchange system with automated analytical instruments. The operational recommendations associated with the use of such HT systems as well as the efficiencies gained (reduction in hands-on time and run time by over 70% and 30%, respectively), which enable practical characterization of an expanded formulation design space, are discussed. To demonstrate that the workflow is fit for purpose, the formulation properties and stability profiles (SEC and CEX) from samples generated by the HT workflow were compared to those processed by ultrafiltration/diafiltration, and the results were shown to be in good agreement. This approach was further applied to two case studies, one focused on a formulation screen that studied the effects of pH and excipient on viscosity and stability, and the other focused on selection of an appropriate viscosity mimic solution for a protein product.
Collapse
Affiliation(s)
- Cindy D Ren
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320.
| | - Wei Qi
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| | - Emily A Wyatt
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| | - Jeffrey Yeary
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| | | | - Michael Berke
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| | - Nitin Rathore
- Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320
| |
Collapse
|
32
|
Maruthamuthu MK, Rudge SR, Ardekani AM, Ladisch MR, Verma MS. Process Analytical Technologies and Data Analytics for the Manufacture of Monoclonal Antibodies. Trends Biotechnol 2020; 38:1169-1186. [PMID: 32839030 PMCID: PMC7442002 DOI: 10.1016/j.tibtech.2020.07.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022]
Abstract
Process analytical technology (PAT) for the manufacture of monoclonal antibodies (mAbs) is defined by an integrated set of advanced and automated methods that analyze the compositions and biophysical properties of cell culture fluids, cell-free product streams, and biotherapeutic molecules that are ultimately formulated into concentrated products. In-line or near-line probes and systems are remarkably well developed, although challenges remain in the determination of the absence of viral loads, detecting microbial or mycoplasma contamination, and applying data-driven deep learning to process monitoring and soft sensors. In this review, we address the current status of PAT for both batch and continuous processing steps and discuss its potential impact on facilitating the continuous manufacture of biotherapeutics.
Collapse
Affiliation(s)
- Murali K. Maruthamuthu
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA,Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Scott R. Rudge
- RMC Pharmaceutical Solutions, Inc., Longmont, CO 80501, USA
| | - Arezoo M. Ardekani
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Michael R. Ladisch
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA,Laboratory of Renewable Resources Engineering, Purdue University, West Lafayette, IN 47907, USA,Correspondence:
| | - Mohit S. Verma
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA,Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA,Correspondence:
| |
Collapse
|
33
|
Deokar V, Sharma A, Mody R, Volety SM. Comparison of Strategies in Development and Manufacturing of Low Viscosity, Ultra-High Concentration Formulation for IgG1 Antibody. J Pharm Sci 2020; 109:3579-3589. [PMID: 32946895 PMCID: PMC7491461 DOI: 10.1016/j.xphs.2020.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/26/2020] [Accepted: 09/08/2020] [Indexed: 12/23/2022]
Abstract
Monoclonal antibodies requiring higher doses for exerting therapeutic effect but having lower stability, are administered as dilute infusions, or as two (low concentration) injections both resulting in reduced patient compliance. Present research summarizes impact of manufacturing conditions on ultra-high concentration (≥150 mg/mL) IgG1 formulation, which can be administered as one subcutaneous injection. IgG1 was concentrated to ~200 mg/mL using tangential flow filtration (TFF). Alternatively, spray dried (SPD) and spray freeze dried (SFD) IgG1, was reconstituted in 30%v/v propylene glycol to form ultra-high concentration (~200 mg/mL) injectable formulation. Reconstituted, SPD and SFD IgG1 formulations, increased viscosity beyond an acceptable range for subcutaneous injections (<20 cP). Formulations developed by reconstitution of SPD IgG1, demonstrated increase in high and low molecular weight impurities, at accelerated and stressed conditions. Whereas, the stability data suggested reconstituted SFD IgG1 was comparable to control IgG1 formulation concentrated by TFF. Also, formulation of IgG1 diafiltered with proline using TFF, reduce viscosity from ~21.9 cP to ~11 cP at 25 °C and had better stability. Thus, conventional TFF technique stands to be one of the preferred methods for manufacturing of ultra-high concentration IgG1 formulations. Additionally, SFD could be an alternative method for long term storage of IgG1 in a dry powder state.
Collapse
Affiliation(s)
- Vaibhav Deokar
- Lupin Limited (Biotechnology Division), A-401, G.O. Square Mall, Sr. No. 249/50, Wakad, Pune 411057, India.
| | - Alok Sharma
- Lupin Limited (Biotechnology Division), A-401, G.O. Square Mall, Sr. No. 249/50, Wakad, Pune 411057, India
| | - Rustom Mody
- Lupin Limited (Biotechnology Division), A-401, G.O. Square Mall, Sr. No. 249/50, Wakad, Pune 411057, India
| | - Subrahmanyam M Volety
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (Deemed University), Manipal 576104, Karnataka, India
| |
Collapse
|
34
|
Brovč EV, Mravljak J, Šink R, Pajk S. Degradation of polysorbates 20 and 80 catalysed by histidine chloride buffer. Eur J Pharm Biopharm 2020; 154:236-245. [PMID: 32693155 DOI: 10.1016/j.ejpb.2020.07.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/01/2020] [Accepted: 07/12/2020] [Indexed: 10/23/2022]
Abstract
Polysorbates are amphiphilic, non-ionic surfactants, and they represent one of the key components of biopharmaceuticals. They serve as stabilisers, and their degradation can cause particle formation, which has been an industry-wide issue over the past decade. To determine the influence of the buffers most frequently used in biopharmaceuticals on polysorbate degradation, an accelerated stability study was carried out using placebo formulations containing 0.02% polysorbates and 20 mM buffers (pH 5.5, 6.5). These included histidine chloride, sodium citrate, sodium succinate and sodium phosphate buffers. The rate of polysorbate degradation was highest in histidine chloride buffer, and therefore we further focused on the mechanism here. The predominant degradation pathway of polysorbates in this buffer was ester hydrolysis, catalysed by the imidazole moiety of the histidine. Interestingly, the presence of therapeutic proteins in the formulations slowed histidine-catalysed degradation of polysorbates in 50% of cases, with negligible degradation seen otherwise. This emphasises the complex nature of the interactions between the components of biopharmaceutical drug products. Nonetheless, there are disadvantages of using histidine chloride buffers in biopharmaceuticals that contain polysorbates. Careful consideration should be given to selection of excipients used in parenteral formulations, whereby compatibility between buffer and surfactant is of key importance.
Collapse
Affiliation(s)
- Ema Valentina Brovč
- University of Ljubljana, Faculty of Pharmacy, The Chair of Pharmaceutical Chemistry, Aškerčeva 7, SI-1000 Ljubljana, Slovenia; Global Drug Development, Technical Research & Development, Novartis, Biologics Technical Development Mengeš, Drug Product Development Biosimilars, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia
| | - Janez Mravljak
- University of Ljubljana, Faculty of Pharmacy, The Chair of Pharmaceutical Chemistry, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
| | - Roman Šink
- Global Drug Development, Technical Research & Development, Novartis, Biologics Technical Development Mengeš, Drug Product Development Biosimilars, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia.
| | - Stane Pajk
- University of Ljubljana, Faculty of Pharmacy, The Chair of Pharmaceutical Chemistry, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
35
|
Brovč EV, Mravljak J, Šink R, Pajk S. Rational design to biologics development: The polysorbates point of view. Int J Pharm 2020; 581:119285. [PMID: 32240804 DOI: 10.1016/j.ijpharm.2020.119285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/29/2022]
Abstract
Formulation development is an essential part of any biopharmaceuticals development programme, and this will affect quality, safety and efficacy of the final drug product. The vast majority of biopharmaceuticals on the market are therapeutic proteins; however, these are less stable compared to conventional pharmaceuticals. To counter aggregation, denaturation and surface adsorption of proteins in solution, surfactants are added to the formulations; however, the choice of the best formulation is a challenge that is faced during formulation development. Polysorbates are the most widely used surfactants in the pharmaceutical industry and are presented in >80% of commercial monoclonal antibody formulations. In this review, we provide a general overview of polysorbates and their issues, and the characteristics that have to be taken into account during formulation development. Degradation of polysorbates, namely by hydrolysis and/or oxidation, is one of the main concerns associated with their use. Furthermore, degradation of polysorbates is determined by formulation composition, pH and storage conditions, therefore underlining the importance and complexity of protein formulation development using polysorbates. A need-based approach should be used for correct selection of excipients in protein formulations that contain polysorbates.
Collapse
Affiliation(s)
- Ema Valentina Brovč
- Chair of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia; Global Drug Development Technical Research & Development, Novartis, Biologics Technical Development Mengeš, Drug Product Development, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia
| | - Janez Mravljak
- Chair of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
| | - Roman Šink
- Global Drug Development Technical Research & Development, Novartis, Biologics Technical Development Mengeš, Drug Product Development, Lek Pharmaceuticals d.d., Kolodvorska 27, SI-1234 Mengeš, Slovenia
| | - Stane Pajk
- Chair of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
36
|
Hu Y, Toth RT, Joshi SB, Esfandiary R, Middaugh CR, Volkin DB, Weis DD. Characterization of Excipient Effects on Reversible Self-Association, Backbone Flexibility, and Solution Properties of an IgG1 Monoclonal Antibody at High Concentrations: Part 2. J Pharm Sci 2020; 109:353-363. [DOI: 10.1016/j.xphs.2019.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/13/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022]
|
37
|
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: 2.3] [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]
|
38
|
Sahin E, Deshmukh S. Challenges and Considerations in Development and Manufacturing of High Concentration Biologics Drug Products. J Pharm Innov 2019. [DOI: 10.1007/s12247-019-09414-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
39
|
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: 3.6] [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
|
40
|
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: 8] [Impact Index Per Article: 1.6] [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
|
41
|
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: 1.8] [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]
|
42
|
Wang W, Ohtake S. Science and art of protein formulation development. Int J Pharm 2019; 568:118505. [PMID: 31306712 DOI: 10.1016/j.ijpharm.2019.118505] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023]
Abstract
Protein pharmaceuticals have become a significant class of marketed drug products and are expected to grow steadily over the next decade. Development of a commercial protein product is, however, a rather complex process. A critical step in this process is formulation development, enabling the final product configuration. A number of challenges still exist in the formulation development process. This review is intended to discuss these challenges, to illustrate the basic formulation development processes, and to compare the options and strategies in practical formulation development.
Collapse
Affiliation(s)
- Wei Wang
- Biological Development, Bayer USA, LLC, 800 Dwight Way, Berkeley, CA 94710, United States.
| | - Satoshi Ohtake
- Pharmaceutical Research and Development, Pfizer Biotherapeutics Pharmaceutical Sciences, Chesterfield, MO 63017, United States
| |
Collapse
|
43
|
Duerr C, Friess W. Antibody-drug conjugates- stability and formulation. Eur J Pharm Biopharm 2019; 139:168-176. [DOI: 10.1016/j.ejpb.2019.03.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 01/01/2023]
|
44
|
Dear BJ, Bollinger JA, Chowdhury A, Hung JJ, Wilks LR, Karouta CA, Ramachandran K, Shay TY, Nieto MP, Sharma A, Cheung JK, Nykypanchuk D, Godfrin PD, Johnston KP, Truskett TM. X-ray Scattering and Coarse-Grained Simulations for Clustering and Interactions of Monoclonal Antibodies at High Concentrations. J Phys Chem B 2019; 123:5274-5290. [DOI: 10.1021/acs.jpcb.9b04478] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Barton J. Dear
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jonathan A. Bollinger
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Center for Integrated Nanotechnologies, Sandia National Laboratories, Albuquerque, New Mexico 87185, United States
| | - Amjad Chowdhury
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jessica J. Hung
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Logan R. Wilks
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Carl A. Karouta
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Kishan Ramachandran
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Tony Y. Shay
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Maria P. Nieto
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Ayush Sharma
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jason K. Cheung
- Biophysical and Biochemical Characterization, Sterile Formulation Sciences, Merck & Co., Inc., Kenilworth, New Jersey 07033 United States
| | - Dmytro Nykypanchuk
- Center for Functional Nanomaterials, Brookhaven National Laboratory, Upton, New York 11973, United States
| | - P. Douglas Godfrin
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Keith P. Johnston
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Thomas M. Truskett
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Physics, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
45
|
Hung JJ, Dear BJ, Karouta CA, Chowdhury AA, Godfrin PD, Bollinger JA, Nieto MP, Wilks LR, Shay TY, Ramachandran K, Sharma A, Cheung JK, Truskett TM, Johnston KP. Protein-Protein Interactions of Highly Concentrated Monoclonal Antibody Solutions via Static Light Scattering and Influence on the Viscosity. J Phys Chem B 2019; 123:739-755. [PMID: 30614707 DOI: 10.1021/acs.jpcb.8b09527] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The ability to design and formulate mAbs to minimize attractive interactions at high concentrations is important for protein processing, stability, and administration, particularly in subcutaneous delivery, where high viscosities are often challenging. The strength of protein-protein interactions (PPIs) of an IgG1 and IgG4 monoclonal antibody (mAb) from low to high concentration was determined by static light scattering (SLS) and used to understand viscosity data. The PPI were tuned using NaCl and five organic ionic co-solutes. The PPI strength was quantified by the normalized structure factor S(0)/ S(0)HS and Kirkwood-Buff integral G22/ G22,HS (HS = hard sphere) determined from the SLS data and also by fits with (1) a spherical Yukawa potential and (2) an interacting hard sphere (IHS) model, which describes attraction in terms of hypothetical oligomers. The IHS model was better able to capture the scattering behavior of the more strongly interacting systems (mAb and/or co-solute) than the spherical Yukawa potential. For each descriptor of PPI, linear correlations were obtained between the viscosity at high concentration (200 mg/mL) and the interaction strengths evaluated both at low (20 mg/mL) and high concentrations (200 mg/mL) for a given mAb. However, the only parameter that provided a correlation across both mAbs was the oligomer mass ratio ( moligomer/ mmonomer+dimer) from the IHS model, indicating the importance of self-association (in addition to the direct influence of the attractive PPI) on the viscosity.
Collapse
Affiliation(s)
- Jessica J Hung
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Barton J Dear
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Carl A Karouta
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Amjad A Chowdhury
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - P Douglas Godfrin
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Jonathan A Bollinger
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States.,Center for Integrated Nanotechnologies , Sandia National Laboratories , Albuquerque , New Mexico 87185 , United States
| | - Maria P Nieto
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Logan R Wilks
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Tony Y Shay
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Kishan Ramachandran
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Ayush Sharma
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Jason K Cheung
- Pharmaceutical Sciences , MRL, Merck & Co., Inc. , Kenilworth , New Jersey 07033 , United States
| | - Thomas M Truskett
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| | - Keith P Johnston
- McKetta Department of Chemical Engineering , The University of Texas at Austin , Austin , Texas 78712 , United States
| |
Collapse
|
46
|
Commentary: New perspectives on protein aggregation during Biopharmaceutical development. Int J Pharm 2018; 552:1-6. [DOI: 10.1016/j.ijpharm.2018.09.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 11/18/2022]
|
47
|
Viola M, Sequeira J, Seiça R, Veiga F, Serra J, Santos AC, Ribeiro AJ. Subcutaneous delivery of monoclonal antibodies: How do we get there? J Control Release 2018; 286:301-314. [DOI: 10.1016/j.jconrel.2018.08.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/29/2022]
|
48
|
Awwad S, Angkawinitwong U. Overview of Antibody Drug Delivery. Pharmaceutics 2018; 10:E83. [PMID: 29973504 PMCID: PMC6161251 DOI: 10.3390/pharmaceutics10030083] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/29/2018] [Accepted: 06/29/2018] [Indexed: 12/11/2022] Open
Abstract
Monoclonal antibodies (mAbs) are one of the most important classes of therapeutic proteins, which are used to treat a wide number of diseases (e.g., oncology, inflammation and autoimmune diseases). Monoclonal antibody technologies are continuing to evolve to develop medicines with increasingly improved safety profiles, with the identification of new drug targets being one key barrier for new antibody development. There are many opportunities for developing antibody formulations for better patient compliance, cost savings and lifecycle management, e.g., subcutaneous formulations. However, mAb-based medicines also have limitations that impact their clinical use; the most prominent challenges are their short pharmacokinetic properties and stability issues during manufacturing, transport and storage that can lead to aggregation and protein denaturation. The development of long acting protein formulations must maintain protein stability and be able to deliver a large enough dose over a prolonged period. Many strategies are being pursued to improve the formulation and dosage forms of antibodies to improve efficacy and to increase the range of applications for the clinical use of mAbs.
Collapse
Affiliation(s)
- Sahar Awwad
- UCL School of Pharmacy, London WC1N 1AX, UK.
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1 V9EL, UK.
| | | |
Collapse
|
49
|
Improving Viscosity and Stability of a Highly Concentrated Monoclonal Antibody Solution with Concentrated Proline. Pharm Res 2018; 35:133. [DOI: 10.1007/s11095-018-2398-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/28/2018] [Indexed: 12/22/2022]
|
50
|
Giannos SA, Kraft ER, Zhao ZY, Merkley KH, Cai J. Formulation Stabilization and Disaggregation of Bevacizumab, Ranibizumab and Aflibercept in Dilute Solutions. Pharm Res 2018; 35:78. [PMID: 29492680 PMCID: PMC5830485 DOI: 10.1007/s11095-018-2368-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/11/2018] [Indexed: 11/16/2022]
Abstract
Purpose Studies were conducted to investigate dilute solutions of the monoclonal antibody (mAb) bevacizumab, mAb fragment ranibizumab and fusion protein aflibercept, develop common procedures for formulation of low concentration mAbs and identify a stabilizing formulation for anti-VEGF mAbs for use in in vitro permeation studies. Methods Excipient substitutions were screened. The most stabilizing formulation was chosen. Standard dilutions of bevacizumab, ranibizumab and aflibercept were prepared in PBS, manufacturer’s formulation, and the new formulation. Analysis was by SE-HPLC and ELISA. Stability, disaggregation and pre-exposure tests were studied. Results When Avastin, Lucentis and Eylea are diluted in PBS or manufacturer’s formulation, there is a 40–50% loss of monomer concentration and drug activity. A formulation containing 0.3% NaCl, 7.5% trehalose, 10 mM arginine and 0.04% Tween 80 at a pH of 6.78 stabilized the mAbs and minimized the drug loss. The formulation also disaggregates mAb aggregation while preserving the activity. Degassing the formulation increases recovery. Conclusions We developed a novel formulation that significantly stabilizes mAbs under unfavorable conditions such as low concentration or body temperature. The formulation allows for tissue permeation experimentation. The formulation also exhibits a disaggregating effect on mAbs, which can be applied to the manufacture/packaging of mAbs and bioassay reagents. Electronic supplementary material The online version of this article (10.1007/s11095-018-2368-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Steven A Giannos
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA.
| | - Edward R Kraft
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA
| | - Zhen-Yang Zhao
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA
| | - Kevin H Merkley
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA
| | - Jiyang Cai
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555, USA
| |
Collapse
|