1
|
Dao HM, Sandoval MA, Cui Z, Williams Iii RO. Reconsidering freeze-induced protein aggregation: Air bubbles as the root cause of ice-water interface stress. Int J Pharm 2024; 665:124723. [PMID: 39299357 DOI: 10.1016/j.ijpharm.2024.124723] [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: 06/20/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Freeze-induced stress causing aggregation of proteins has typically been primarily attributed to the ice-water interface. However, we hypothesize that the underlying observed and perceived detrimental effect of ice is, to some extent, attributed to air bubbles expelled from ice crystal lattices or to nanobubbles existing prior to freezing. The reduction of dissolved air was achieved via a deaeration process by placing samples in a reduced pressure chamber, while the reduction of nanobubbles was achieved by filtering samples via a syringe filter. The results showed that the reduction of both dissolved air molecules and stable colloidal nanobubbles in a bovine IgG solution prior to freezing led to a significant decrease in aggregation after thawing compared to untreated samples (∼6,000 vs. ∼ 40,000 particles/mL at a freezing rate of 100 K/s, respectively). The deaeration-filtration treatment works additively with cryoprotectants such as trehalose, further reducing the freeze-induced aggregation of IgG. The results also demonstrated that air-water interfacial aggregation of IgG in bulk liquid samples is a time-dependent process. The number of IgG subvisible particles increased with time and temperature, suggesting that random collisions of denatured molecules promoted the formation of aggregates with spherical morphology. In contrast, the IgG subvisible count after freeze-thawing had already reached its nominal value, suggesting a time-independent process where denatured protein molecules were compressed between ice crystals into filament-like aggregates. In summary, the findings shift the current paradigm from ice crystals being the main destabilizing factor during freezing to air bubbles, although the two are intertwined. From a translational aspect, this study underscores the value of deaeration-filtration as an essential supplemental process that can be applied in addition to formulation approaches such as the use of cryoprotectants to further reduce freezing stress on proteins and increase their stability.
Collapse
Affiliation(s)
- Huy M Dao
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Michael A Sandoval
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Zhengrong Cui
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Robert O Williams Iii
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
2
|
Choi SM, Lee JH, Ko S, Hong SS, Jin HE. Mechanism of Action and Pharmacokinetics of Approved Bispecific Antibodies. Biomol Ther (Seoul) 2024; 32:708-722. [PMID: 39448393 PMCID: PMC11535297 DOI: 10.4062/biomolther.2024.146] [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: 08/21/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Bispecific antibodies represent a significant advancement in therapeutic antibody engineering, offering the ability to simultaneously target two distinct antigens. This dual-targeting capability enhances therapeutic efficacy, especially in complex diseases, such as cancer and autoimmune disorders, where drug resistance and incomplete target coverage are prevalent challenges. Bispecific antibodies facilitate immune cell engagement and disrupt multiple signaling pathways, providing a more comprehensive treatment approach than traditional monoclonal antibodies. However, the intricate structure of bispecific antibodies introduces unique pharmacokinetic challenges, including issues related to their absorption, distribution, metabolism, and excretion, which can significantly affect their efficacy and safety. This review provides an in-depth analysis of the structural design, mechanisms of action, and pharmacokinetics of the currently approved bispecific antibodies. It also highlights the engineering innovations that have been implemented to overcome these challenges, such as Fc modifications and advanced dimerization techniques, which enhance the stability and half-life of bispecific antibodies. Significant progress has been made in bispecific antibody technology; however, further research is necessary to broaden their clinical applications, enhance their safety profiles, and optimize their incorporation into combination therapies. Continuous advancements in this field are expected to enable bispecific antibodies to provide more precise and effective therapeutic strategies for a range of complex diseases, ultimately improving patient outcomes and advancing precision medicine.
Collapse
Affiliation(s)
- Seong Min Choi
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Soyeon Ko
- Department of Biomedical Sciences, College of Medicine & Program in Biomedicals Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine & Program in Biomedicals Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Hyo-Eon Jin
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
3
|
Asmani AZA, Zainuddin AFF, Azmi Murad NA, Mohd Darwis NH, Suhaimi NS, Zaini E, Taher M, Susanti D, Khotib J. Immunogenicity of monoclonal antibody: Causes, consequences, and control strategies. Pathol Res Pract 2024; 263:155627. [PMID: 39357185 DOI: 10.1016/j.prp.2024.155627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/14/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Antibody-based treatment was first used in 1891 for the treatment of diphtheria. Since then, monoclonal antibodies (mAbs) have been developed to treat many diseases such as cancer and act as vaccines. However, murine-derived therapeutic mAbs were found to be highly immunogenic, and caused anti-drug antibodies (ADAs) reaction, reducing their efficacy and causing severe infusion reactions. Fully human, humanised, and chimeric antibodies were then introduced for better therapeutic efficacy. With the introduction of immune response associated with mAbs immunogenicity. This review explores the immunogenicity of mAbs, its mechanism, contributing factors, and its impact on therapeutic efficacy. It also discusses immunogenicity assessment for preclinical studies and strategies for minimising immunogenicity for effective therapeutic treatment in various diseases. Finally, predicting immunogenicity in drug development is essential for selecting top drug candidates. A lot of methods can be implemented by the researchers and developers to reduce the development of ADAs while simultaneously minimising the immunogenicity reaction of mAbs.
Collapse
Affiliation(s)
- Ahmad Zafran Amin Asmani
- Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Ahmad Faris Fahmi Zainuddin
- Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Nadhirah Ahmad Azmi Murad
- Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Nur Hidayati Mohd Darwis
- Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Nur Suhaida Suhaimi
- Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia
| | - Erizal Zaini
- Faculty of Pharmacy, Universitas Andalas, Padang 25175, Indonesia
| | - Muhammad Taher
- Faculty of Pharmacy, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia.
| | - Deny Susanti
- Department of Chemistry, Faculty of Science, International Islamic University Malaysia, Jalan Sultan Ahmad Shah, Kuantan, Pahang 25200, Malaysia.
| | - Junaidi Khotib
- Department of Pharmacy Practice, Faculty of Pharmacy, Airlangga University, Surabaya 60115, Indonesia.
| |
Collapse
|
4
|
Cohrs M, Clottens N, Ramaut P, Braeckmans K, De Smedt S, Bauters T, Svilenov HL. Impact of pneumatic tube transportation on the aggregation of monoclonal antibodies in clinical practice. Eur J Pharm Sci 2024:106952. [PMID: 39481661 DOI: 10.1016/j.ejps.2024.106952] [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: 08/19/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Postproduction handling and in-hospital transportation of antibody drugs cause mechanical stress, including interfacial and shear stress, that can induce antibody unfolding and aggregation. The handling practices differ significantly between hospitals and the impact on protein stability is unknown. For example, the mechanical stress caused by transport via pneumatic tube systems (PTS) on therapeutic antibody aggregation is a potential safety and quality gap. The aim of this study was to investigate whether mechanical stress and PTS transportation in a hospital cause aggregation of five commonly used antibody drugs diluted in infusion bags. Orthogonal analytical methods showed that the handling and PTS transportation in this hospital did not cause aggregation of the investigated mAbs. The absence of aggregation could be explained by the reduction of interfacial stress due to headspace removal from the infusion bags and a mechanical sensor indicated that there was also only a moderate amount of mechanical stress caused by transportation with this particular PTS. Although this case study focuses on five antibody drugs and the practices in one hospital, the work demonstrates how to evaluate whether other handling and transportation practices cause significant mechanical stress that could compromise the quality and safety of antibody drugs.
Collapse
Affiliation(s)
- Michaela Cohrs
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Nele Clottens
- Pharmacy Department, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Pieter Ramaut
- Pharmacy Department, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Tiene Bauters
- Pharmacy Department, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium,.
| | - Hristo L Svilenov
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Biopharmaceutical Technology, TUM School of Life Sciences, Technical University of Munich, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany.
| |
Collapse
|
5
|
Cohen JR, Joubert MK, Tabassum S, Capili A, Carreon J, Xiang C, Prabhu S, Merlo A, Mytych D, Dolan DG, Kouda R. Experimental Validation of a Parenteral Permitted Daily Exposure Value for Cleaning-Induced Degradants from Recombinant Therapeutic Proteins with In Vitro Immunogenicity Assays. J Pharm Sci 2024:S0022-3549(24)00490-8. [PMID: 39490658 DOI: 10.1016/j.xphs.2024.10.041] [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: 06/21/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Multiproduct manufacturing of biotherapeutic proteins generate cleaning-induced protein degradants because of extreme pH and temperature conditions during the cleaning process. Cleaning Acceptance limits are calculated based on the maximum allowable carryover (MAC) assessment of the previously manufactured active pharmaceutical ingredient (API) - or drug product - based on the permitted daily exposure (PDE) of the previously manufactured API into the dose of subsequent product. In this study, we tested a previously determined PDE value for cleaning-induced protein degradants of 650 µg/dose. A bench-scale cleaning method was used to generate cleaning induced degradants from both a half-life extension (HLE) BiTE® molecule and a mAb product. For this investigation degradants of HLE BiTE®-A and mAb-1 were characterized either alone or degradants of HLE BiTE®-A and mAb-1 spiked into mAb-1 at 650 µg. These samples were characterized by endotoxin testing, size exclusion chromatography (SEC), light obscuration by HIAC, and micro-fluidic imaging (MFI). These results suggest that significant degradation of the molecule occurs because of the cleaning procedure, and it is no longer in the intact form or active state. The biological impact was assessed using a cell line assay to assess immune activation, and a human Peripheral Blood Mononuclear Cell (PBMC) assay to assess T cell activation, T cell proliferation, and cytokine release after 20 hours and 7 days. Findings from the various in vitro cell-based assays suggest that the presence of 650 µg of carryover of degradants either alone or spiked into the same or a cross-product do not increase immunogenicity risk in cell-based assays - suggesting that the current PDE of 650 µg/dose for cleaning-induced degradant carryover does not have a risk of immunogenicity in patients.
Collapse
Affiliation(s)
- Joseph R Cohen
- The Department of Process Development, Amgen Inc., Thousand Oaks, CA 91320.
| | - Marisa K Joubert
- The Department of Process Development, Amgen Inc., Thousand Oaks, CA 91320
| | - Syeda Tabassum
- The Department of Process Development, Amgen Inc., Thousand Oaks, CA 91320
| | - Allyson Capili
- The Department of Process Development, Amgen Inc., Thousand Oaks, CA 91320
| | - Julia Carreon
- The Department of Process Development, Amgen Inc., Thousand Oaks, CA 91320
| | - Cathie Xiang
- The Department of Process Development, Amgen Inc., Thousand Oaks, CA 91320
| | - Siddharth Prabhu
- The Department of Process Development, Amgen Inc., Thousand Oaks, CA 91320
| | - Anthony Merlo
- The Department of Process Development, Amgen Inc., Thousand Oaks, CA 91320
| | - Dan Mytych
- The Department of Clinical Immunology, Amgen Inc., Thousand Oaks, CA 91320
| | - David G Dolan
- The Department of Environmental Health and Safety, Amgen Inc., Thousand Oaks, CA 91320
| | - Ram Kouda
- The Department of Process Development, Amgen Inc., Thousand Oaks, CA 91320.
| |
Collapse
|
6
|
Martins Fraga R, Beretta M, Pinto JF, Spoerk M, Zupančič O, Pinto JT, Paudel A. Effect of processing and formulation factors on Catalase activity in tablets. Int J Pharm 2024; 664:124626. [PMID: 39208952 DOI: 10.1016/j.ijpharm.2024.124626] [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: 06/14/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
The manufacturing of tablets containing biologics exposes the biologics to thermal and shear stresses, which are likely to induce structural changes (e.g., aggregation and denaturation), leading to the loss of their activity. Saccharides often act as stabilizers of proteins in formulations, yet their stabilizing ability throughout solid oral dosage processing, such as tableting, has been barely studied. This work aimed to investigate the effects of formulation and process (tableting and spray-drying) variables on catalase tablets containing dextran, mannitol, and trehalose as potential stabilizers. Non-spray-dried and spray-dried formulations were prepared and tableted (100, 200, and 400 MPa). The enzymatic activity, number of aggregates, reflecting protein aggregation and structure modifications were studied. A principal component analysis was performed to reveal underlying correlations. It was found that tableting and spray-drying had a notable negative effect on the activity and number of aggregates formed in catalase formulations. Overall, dextran and mannitol failed to preserve the catalase activity in any unit operation studied. On the other hand, trehalose was found to preserve the activity during spray-drying but not necessarily during tableting. The study demonstrated that formulation and process variables must be considered and optimized together to preserve the characteristics of catalase throughout processing.
Collapse
Affiliation(s)
- Rúben Martins Fraga
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Michela Beretta
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - João F Pinto
- iMed.UL - Faculdade de Farmácia da Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Martin Spoerk
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria; Institute of Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13, 8010 Graz, Austria
| | - Ožbej Zupančič
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Joana T Pinto
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Amrit Paudel
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria; Institute of Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13, 8010 Graz, Austria.
| |
Collapse
|
7
|
Shrestha KR, Kim S, Jo A, Ragothaman M, Yoo SY. In vivo safety evaluation and tracing of arginylglycylaspartic acid-engineered phage nanofiber in murine model. J Mater Chem B 2024; 12:10258-10271. [PMID: 39300937 DOI: 10.1039/d4tb00823e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The engineered phage YSY184, mimicking the extracellular matrix nanofiber, effectively promotes stem cell differentiation and angiogenesis. This study evaluated its safety in a mouse model, monitoring weight, immunogenicity, spleen immune responses, and macrophage infiltration. Rapid clearance of YSY184 was observed, with peak tissue presence within three hours, significantly reduced by 24 hours, and negligible after one month. No adverse physiological or pathological effects were detected post-administration, affirming YSY184's safety and underscore its potential for therapeutic use, warranting further clinical exploration.
Collapse
Affiliation(s)
- Kshitiz Raj Shrestha
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| | - Sehoon Kim
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| | - Anna Jo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| | - Murali Ragothaman
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| | - So Young Yoo
- Institute of Nanobio Convergence, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
8
|
Jordan JS, Harper CC, Williams ER. High-Throughput Single-Particle Characterization of Aggregation Pathways and the Effects of Inhibitors for Large (Megadalton) Protein Oligomers. Anal Chem 2024. [PMID: 39394988 DOI: 10.1021/acs.analchem.4c04669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Protein aggregation is involved in many human diseases, but characterizing the sizes and shapes of intermediate oligomers (∼10-100 nm) that are important to the formation of macroscale aggregates like amyloid fibrils is a significant analytical challenge. Here, charge detection mass spectrometry (CDMS) is used to characterize individual conformational states of bovine serum albumin oligomers with up to ∼225 molecules (15 MDa). Elongated, partially folded, and globular conformational families for each oligomer can be readily distinguished based on the extent of charging. The abundances of individual conformers vary with changes in the monomer concentration or by adding aggregation inhibitors, such as SDS, heparin, or MgCl2. These results show the potential of CDMS for investigating intermediate oligomers in protein aggregation processes that are important for understanding aggregate formation and inhibition mechanisms and could accelerate formulation buffer development to prevent the aggregation of biotherapeutics.
Collapse
Affiliation(s)
- Jacob S Jordan
- Department of Chemistry, University of California, Berkeley, California 94720-1460, United States
| | - Conner C Harper
- Department of Chemistry, University of California, Berkeley, California 94720-1460, United States
| | - Evan R Williams
- Department of Chemistry, University of California, Berkeley, California 94720-1460, United States
| |
Collapse
|
9
|
Velankar KY, Gawalt ES, Wen Y, Meng WS. Pharmaceutical proteins at the interfaces and the role of albumin. Biotechnol Prog 2024; 40:e3474. [PMID: 38647437 DOI: 10.1002/btpr.3474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/15/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
A critical measure of the quality of pharmaceutical proteins is the preservation of native conformations of the active pharmaceutical ingredients. Denaturation of the active proteins in any step before administration into patients could lead to loss of potency and/or aggregation, which is associated with an increased risk of immunogenicity of the products. Interfacial stress enhances protein instability as their adsorption to the air-liquid and liquid-solid interfaces are implicated in the formation of denatured proteins and aggregates. While excipients in protein formulations have been employed to reduce the risk of aggregation, the roles of albumin as a stabilizer have not been reviewed from practical and theoretical standpoints. The amphiphilic nature of albumin makes it accumulate at the interfaces. In this review, we aim to bridge the knowledge gap between interfacial instability and the influence of albumin as a surface-active excipient in the context of reducing the immunogenicity risk of protein formulations.
Collapse
Affiliation(s)
- Ketki Y Velankar
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Ellen S Gawalt
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
Islam SN, Arif Z, Badar A, Moinuddin, Khan MA, Alam K. Glycoxidation of mammalian whole histone generates highly immunogenic aggregates: Sera of SLE patients contain autoantibodies against aggregates. Scand J Immunol 2024; 100:e13389. [PMID: 38816907 DOI: 10.1111/sji.13389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 04/15/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
Non-enzymatic glycation and oxidation of self-proteins, causing formation and accumulation of advanced glycation end products (AGEs), have been reported in an array of pathologies, including systemic lupus erythematosus (SLE). Such modifications may generate neo-epitopes, break immunological tolerance, and induce antibody response. In this study, we have first analysed the structural modifications of whole histone in the presence of deoxyribose followed by oxidation with hydroxyl radicals. Changes in the secondary and tertiary structure of the whole histone were determined by spectroscopic techniques and biochemical assays. Fluorescence spectroscopy and UPLC-MS showed the generation of AGEs such as carboxymethyl lysine and pentosidine, while DLS and TEM indicated the presence of amorphous AGE-aggregates. Moreover, rabbits immunized with these histone-AGEs exhibited enhanced immunogenicity and ELISA and western immunoblot of IgG antibodies from SLE patients' sera showed a significantly higher specificity towards modified histone-AGEs than the native histone.
Collapse
Affiliation(s)
- Shireen Naaz Islam
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| | - Zarina Arif
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| | - Asim Badar
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| | - Moinuddin
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| | - Md Asad Khan
- Department of Biochemistry, Faculty of Dentistry, Jamia Millia Islamia, New Delhi, India
| | - Khursheed Alam
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| |
Collapse
|
11
|
Hasan HJ, Ghareeb MM. Optimizing Desolvation Conditions for Glutathione-Cross-Linked Bovine Serum Albumin Nanoparticles: Implication for Intravenous Drug Delivery. Cureus 2024; 16:e69514. [PMID: 39416524 PMCID: PMC11481410 DOI: 10.7759/cureus.69514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2024] [Indexed: 10/19/2024] Open
Abstract
INTRODUCTION Protein-based nanocarriers, particularly albumin nanoparticles (NPs), offer numerous advantages when compared to other nanomaterials. These carriers are characterized by biocompatibility, biodegradability, reduced immunogenicity, and decreased cytotoxicity. Moreover, proteins possess an inherent ability to target tumor cells directly or indirectly. AIM This study aims to investigate the impact of various organic solvents on the characteristics of synthesized bovine serum albumin NPs (BSA NPs). METHOD BSA NPs were produced using methanol, acetone, ethanol, dimethylsulfoxide (DMSO), and acetonitrile through the desolvation technique to achieve particles of acceptable size. Dynamic light scattering (DLS), blood compatibility assays, polyacrylamide gel electrophoresis (PAGE), and size exclusion chromatography (SEC) were employed to elucidate the properties of the generated NPs. The cytotoxicity of BSA NPs prepared under different conditions was assessed using Michigan Cancer Foundation - Mammary Adenocarcinoma - Breast Cancer 231 cells (MDA-MB-231 cells). RESULTS The particle size of the synthesized NPs varied based on the organic solvent utilized, with the smallest size of 114.2 nm observed with methanol. Blood compatibility results indicated no abnormal interactions between BSA NPs and blood components. PAGE analysis revealed a strong band near 72 kDa for untreated BSA and BSA treated with all organic solvents. In SEC, the retention time of native albumin was 6.65 min, while the average retention times of the prepared BSA NPs ranged from 5.14 to 5.21 min, showing similarity to the native protein. Except for NPs produced with methanol and acetonitrile, cytotoxicity testing on MDA-MB-231 cells demonstrated no significant harmful effects at various concentrations (0-500 μg/mL). CONCLUSION The choice of desolvating agent significantly influences the size of BSA NPs. Various factors, such as solvent characteristics like hydrogen bonds, polarity, dielectric constant, and functional groups, can affect the particle size and structure of BSA NPs. The compatibility of cross-linked BSA NPs with blood components suggests their potential for intravenous drug delivery applications.
Collapse
Affiliation(s)
- Hamid J Hasan
- Department of Pharmaceutics, College of Pharmacy, University of Baghdad, Oncology Teaching Hospital - Medical City, Baghdad, IRQ
| | - Mowafaq M Ghareeb
- Department of Pharmaceutics, College of Pharmacy, University of Baghdad, Baghdad, IRQ
| |
Collapse
|
12
|
Jordan JS, Harper CC, Zhang F, Kofman E, Li M, Sathiyamoorthy K, Zaragoza JP, Fayadat-Dilman L, Williams ER. Charge Detection Mass Spectrometry Reveals Conformational Heterogeneity in Megadalton-Sized Monoclonal Antibody Aggregates. J Am Chem Soc 2024; 146:23297-23305. [PMID: 39110484 DOI: 10.1021/jacs.4c05885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Aggregation of protein-based therapeutics can occur during development, production, or storage and can lead to loss of efficacy and potential toxicity. Native mass spectrometry of a covalently linked pentameric monoclonal antibody complex with a mass of ∼800 kDa reveals several distinct conformations, smaller complexes, and abundant higher-order aggregates of the pentameric species. Charge detection mass spectrometry (CDMS) reveals individual oligomers up to the pentamer mAb trimer (15 individual mAb molecules; ∼2.4 MDa) whereas intermediate aggregates composed of 6-9 mAb molecules and aggregates larger than the pentameric dimer (1.6 MDa) were not detected/resolved by standard mass spectrometry, size exclusion chromatography (SEC), capillary electrophoresis (CE-SDS), or by mass photometry. Conventional quadrupole time-of-flight mass spectrometry (QTOF MS), mass photometry, SEC, and CE-SDS did not resolve partially or more fully unfolded conformations of each oligomer that were readily identified using CDMS by their significantly higher extents of charging. Trends in the charge-state distributions of individual oligomers provides detailed insight into how the structures of compact and elongated mAb aggregates change as a function of aggregate size. These results demonstrate the advantages of CDMS for obtaining accurate masses and information about the conformations of large antibody aggregates despite extensive overlapping m/z values. These results open up the ability to investigate structural changes that occur in small, soluble oligomers during the earliest stages of aggregation for antibodies or other proteins.
Collapse
Affiliation(s)
- Jacob S Jordan
- Department of Chemistry, University of California, Berkeley, California 94720-1460, United States
| | - Conner C Harper
- Department of Chemistry, University of California, Berkeley, California 94720-1460, United States
| | - Fan Zhang
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., 213 E Grand Ave., South San Francisco, California 94080, United States
| | - Esther Kofman
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., 213 E Grand Ave., South San Francisco, California 94080, United States
| | - Mandy Li
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., 213 E Grand Ave., South San Francisco, California 94080, United States
| | - Karthik Sathiyamoorthy
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., 213 E Grand Ave., South San Francisco, California 94080, United States
| | - Jan Paulo Zaragoza
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., 213 E Grand Ave., South San Francisco, California 94080, United States
| | - Laurence Fayadat-Dilman
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., 213 E Grand Ave., South San Francisco, California 94080, United States
| | - Evan R Williams
- Department of Chemistry, University of California, Berkeley, California 94720-1460, United States
| |
Collapse
|
13
|
Kamelnia R, Ahmadi-Hamedani M, Darroudi M, Kamelnia E. Improving the stability of insulin through effective chemical modifications: A Comprehensive review. Int J Pharm 2024; 661:124399. [PMID: 38944170 DOI: 10.1016/j.ijpharm.2024.124399] [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: 03/01/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Insulin, an essential peptide hormone, conjointly regulates blood glucose levels by its receptor and it is used as vital drug to treat diabetes. This therapeutic hormone may undergo different chemical modifications during industrial processes, pharmaceutical formulation, and through its endogenous storage in the pancreatic β-cells. Insulin is highly sensitive to environmental stresses and readily undergoes structural changes, being also able to unfold and aggregate in physiological conditions. Even; small changes altering the structural integrity of insulin may have significant impacts on its biological efficacy to its physiological and pharmacological activities. Insulin analogs have been engineered to achieve modified properties, such as improved stability, solubility, and pharmacokinetics, while preserving the molecular pharmacology of insulin. The casually or purposively strategies of chemical modifications of insulin occurred to improve its therapeutic and pharmaceutical properties. Knowing the effects of chemical modification, formation of aggregates, and nanoparticles on protein can be a new look at the production of protein analogues drugs and its application in living system. The project focused on effects of chemical modifications and nanoparticles on the structure, stability, aggregation and their results in effective drug delivery system, biological activity, and pharmacological properties of insulin. The future challenge in biotechnology and pharmacokinetic arises from the complexity of biopharmaceuticals, which are often molecular structures that require formulation and delivery strategies to ensure their efficacy and safety.
Collapse
Affiliation(s)
- Reyhane Kamelnia
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran
| | - Mahmood Ahmadi-Hamedani
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran.
| | - Majid Darroudi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elahe Kamelnia
- Department of biology, Faculty of sciences, Mashhad branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
14
|
Oluwole SA, Weldu WD, Jayaraman K, Barnard KA, Agatemor C. Design Principles for Immunomodulatory Biomaterials. ACS APPLIED BIO MATERIALS 2024. [PMID: 38922334 DOI: 10.1021/acsabm.4c00537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
The immune system is imperative to the survival of all biological organisms. A functional immune system protects the organism by detecting and eliminating foreign and host aberrant molecules. Conversely, a dysfunctional immune system characterized by an overactive or weakened immune system causes life-threatening autoimmune or immunodeficiency diseases. Therefore, a critical need exists to develop technologies that regulate the immune system to ensure homeostasis or treat several diseases. Accumulating evidence shows that biomaterials─artificial materials (polymers, metals, ceramics, or engineered cells and tissues) that interact with biological systems─can trigger immune responses, offering a materials science-based strategy to modulate the immune system. This Review discusses the expanding frontiers of biomaterial-based immunomodulation, focusing on principles for designing these materials. This Review also presents examples of immunomodulatory biomaterials, which include polymers and metal- and carbon-based nanomaterials, capable of regulating the innate and adaptive immune systems.
Collapse
Affiliation(s)
- Samuel Abidemi Oluwole
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
| | - Welday Desta Weldu
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
| | - Keerthana Jayaraman
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
| | - Kelsie Amanda Barnard
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
| | - Christian Agatemor
- Department of Chemistry, University of Miami, Coral Gables, Florida 33124, United States
- Department of Biology, University of Miami, Coral Gables, Florida 33124, United States
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, Florida 33136, United States
| |
Collapse
|
15
|
Liu F, Hutchinson R. Visible particles in parenteral drug products: A review of current safety assessment practice. Curr Res Toxicol 2024; 7:100175. [PMID: 38975062 PMCID: PMC11223083 DOI: 10.1016/j.crtox.2024.100175] [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: 03/29/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024] Open
Abstract
Parenteral drug products (PDPs) are administered extensively to treat various diseases. Product quality plays a critical role in ensuring patient safety and product efficacy. One important quality challenge is the contamination of particles in PDPs. Particle presence in PDPs represents potential safety risk to patients. Differential guidance and practice have been in place for visible (VPs) and subvisible particles (SVPs) in PDPs. For SVPs, the amount limits have been harmonized in multiple Pharmacopeias. The pharmaceutical industry follows the guided limits for regulatory and quality compliance. However, for VPs, no such acceptable limit has been set. This results in not only quality but also safety challenges for manufacturers and drug developers in managing and evaluating VPs. It is important to understand the potential safety risk of VPs so these can be weighed against the benefit of the PDPs. To evaluate their potential risk(s), it is necessary to understand their nature, origin, frequency of their occurrence, safety risk, the risk mitigation measures, and the method to evaluate their safety. The current paper reviews the critical literature on these aspects and provides insight into considerations when performing safety assessment and managing the risk(s) for VPs in PDPs.
Collapse
Affiliation(s)
- Frank Liu
- Safe Product Services LLC, Pittsfield, MA, USA
| | | |
Collapse
|
16
|
Fernandez-Mendivil C, Kinsella NM, Ebbers HC. A Retrospective Analysis of the Potential Impact of Differences in Aggregates on Clinical Immunogenicity of Biosimilars and their Reference Products. Clin Pharmacol Ther 2024; 115:1122-1131. [PMID: 38284504 DOI: 10.1002/cpt.3180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/03/2024] [Indexed: 01/30/2024]
Abstract
Aggregates, in particular high molecular weight species (HMWs), have been linked to increased immunogenicity. The current understanding on the impact of HMWs is mainly based on in vitro and nonclinical studies and there are only limited data available associating differences in HMWs in marketed monoclonal antibodies (mAbs) to clinical outcomes. Biosimilars offer a unique opportunity to study the potential association between quality parameters and clinical outcomes. We performed a retrospective evaluation to investigate the association between HMW content and reported antidrug-antibody (ADA) incidence in 30 full-length biosimilar mAbs approved in the European Union and the United States. Information for HMW content and ADA incidence were collected from publicly available sources. Differences in HMW content between biosimilars and their reference products (RPs) ranged from -0.75 to 1.65% with slightly higher differences observed in antineoplastic products as compared with immunosuppressant products. The difference in the ADA incidence between the RP and the biosimilar for the programs studied ranged from -11.0 to 18.5%. No association was observed between differences in HMW content and reported ADA incidence, in neither phase I nor phase III studies. Our results show that the limited differences in the content of HMWs between marketed biosimilars and reference mAbs were not associated with differences in reported immunogenicity, determined as incidence of ADAs and neutralizing ADAs in comparative clinical studies.
Collapse
|
17
|
Gorantla A, Hall JTVE, Troidle A, Janjic JM. Biomaterials for Protein Delivery: Opportunities and Challenges to Clinical Translation. MICROMACHINES 2024; 15:533. [PMID: 38675344 PMCID: PMC11052476 DOI: 10.3390/mi15040533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
The development of biomaterials for protein delivery is an emerging field that spans materials science, bioengineering, and medicine. In this review, we highlight the immense potential of protein-delivering biomaterials as therapeutic options and discuss the multifaceted challenges inherent to the field. We address current advancements and approaches in protein delivery that leverage stimuli-responsive materials, harness advanced fabrication techniques like 3D printing, and integrate nanotechnologies for greater targeting and improved stability, efficacy, and tolerability profiles. We also discuss the demand for highly complex delivery systems to maintain structural integrity and functionality of the protein payload. Finally, we discuss barriers to clinical translation, such as biocompatibility, immunogenicity, achieving reliable controlled release, efficient and targeted delivery, stability issues, scalability of production, and navigating the regulatory landscape for such materials. Overall, this review summarizes insights from a survey of the current literature and sheds light on the interplay between innovation and the practical implementation of biomaterials for protein delivery.
Collapse
Affiliation(s)
- Amogh Gorantla
- Department of Engineering, Wake Forest University, Winston-Salem, NC 27109, USA;
| | | | | | - Jelena M. Janjic
- School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA;
| |
Collapse
|
18
|
Cohen JR, Brych SR, Prabhu S, Bi V, Elbaradei A, Tokuda JM, Xiang C, Hokom M, Cui X, Ly C, Amos N, Sun J, Calamba D, Herskovitz J, Capili A, Nourbakhsh K, Merlo A, Carreon J, Wypych J, Narhi LO, Jawa V, Joubert MK. A High Threshold of Biotherapeutic Aggregate Numbers is Needed to Induce an Immunogenic Response In Vitro, In Vivo, and in the Clinic. Pharm Res 2024; 41:651-672. [PMID: 38519817 DOI: 10.1007/s11095-024-03678-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/15/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND AND PURPOSE There is concern that subvisible aggregates in biotherapeutic drug products pose a risk to patient safety. We investigated the threshold of biotherapeutic aggregates needed to induce immunogenic responses. METHODS AND RESULTS Highly aggregated samples were tested in cell-based assays and induced cellular responses in a manner that depended on the number of particles. The threshold of immune activation varied by disease state (cancer, rheumatoid arthritis, allergy), concomitant therapies, and particle number. Compared to healthy donors, disease state patients showed an equal or lower response at the late phase (7 days), suggesting they may not have a higher risk of responding to aggregates. Xeno-het mice were used to assess the threshold of immune activation in vivo. Although highly aggregated samples (~ 1,600,000 particles/mL) induced a weak and transient immunogenic response in mice, a 100-fold dilution of this sample (~ 16,000 particles/mL) did not induce immunogenicity. To confirm this result, subvisible particles (up to ~ 18,000 particles/mL, containing aggregates and silicone oil droplets) produced under representative administration practices (created upon infusion of a drug product through an IV catheter) did not induce a response in cell-based assays or appear to increase the rate of adverse events or immunogenicity during phase 3 clinical trials. CONCLUSION The ability of biotherapeutic aggregates to elicit an immune response in vitro, in vivo, and in the clinic depends on high numbers of particles. This suggests that there is a high threshold for aggregates to induce an immunogenic response which is well beyond that seen in standard biotherapeutic drug products.
Collapse
Affiliation(s)
- Joseph R Cohen
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA.
| | - Stephen R Brych
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Siddharth Prabhu
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Vivian Bi
- The Department of Biosimilars, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Ahmed Elbaradei
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Joshua M Tokuda
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Cathie Xiang
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Martha Hokom
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of BioAnalytical Sciences, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Xiaohong Cui
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Claudia Ly
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Nathan Amos
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Jilin Sun
- Translational Safety and Bioanalytical Sciences, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Dominador Calamba
- Translational Safety and Bioanalytical Sciences, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Jonathan Herskovitz
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Allyson Capili
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Kimya Nourbakhsh
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Anthony Merlo
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Julia Carreon
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Jette Wypych
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Linda O Narhi
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Vibha Jawa
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | - Marisa K Joubert
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA.
| |
Collapse
|
19
|
Migliaccio AR. Erythropoietin: A Personal Alice in Wonderland Trip in the Shadow of the Giants. Biomolecules 2024; 14:408. [PMID: 38672425 PMCID: PMC11047939 DOI: 10.3390/biom14040408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
The identification of the hormone erythropoietin (EPO), which regulates red blood cell production, and its development into a pharmaceutical-grade product to treat anemia has been not only a herculean task but it has also been the first of its kind. As with all the successes, it had "winners" and "losers", but its history is mostly told by the winners who, over the years, have published excellent scientific and divulgate summaries on the subject, some of which are cited in this review. In addition, "success" is also due to the superb and dedicated work of numerous "crew" members, who often are under-represented and under-recognized when the story is told and often have several "dark sides" that are not told in the polished context of most reviews, but which raised the need for the development of the current legislation on biotherapeutics. Although I was marginally involved in the clinical development of erythropoietin, I have known on a personal basis most, if not all, the protagonists of the saga and had multiple opportunities to talk with them on the drive that supported their activities. Here, I will summarize the major steps in the development of erythropoietin as the first bioproduct to enter the clinic. Some of the "dark sides" will also be mentioned to emphasize what a beautiful achievement of humankind this process has been and how the various unforeseen challenges that emerged were progressively addressed in the interest of science and of the patient's wellbeing.
Collapse
|
20
|
Hobson AD. The medicinal chemistry evolution of antibody-drug conjugates. RSC Med Chem 2024; 15:809-831. [PMID: 38516594 PMCID: PMC10953486 DOI: 10.1039/d3md00674c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
Antibody-drug conjugates (ADCs) comprise 3 components of wildly differing sizes: antibody (150 000 Da), linker (typically <500 Da) and payload (typically <500 Da). While the drug-linker makes up only a small percent of the ADC it has a disproportionately massive impact on all aspects of the ADC. Replacing maleimide with bromoacetamide (BrAc) affords stable attachment of the linker to the antibody cysteine, supports total flexibility for linker design and affords a more homogenous ADC. Optimisation of the protease cleavable dipeptide reduces aggregation, facilitates moderation of the physicochemical properties of the ADC and enables long-term stability to facilitate subcutaneous self-administration. Payloads are designed specifically to afford the optimal ADC. Structural information and SAR guide design to improve both potency and selectivity to the small molecule target improving the therapeutic index of resulting ADCs. Minimising the solvent exposed hydrophobic surface area improves the drug-like properties of the ADC, the realisation that the attachment heteroatom can be more than just the site for linker attachment as it can also drive potency and selectivity of the payload and the adoption of a prodrug strategy at project initiation are key areas that medicinal chemistry drives. For an optimal ADC the symbiotic relationship of the three structurally disparate components requires they all function in unison and medicinal chemistry has a huge role to ensure this happens.
Collapse
Affiliation(s)
- Adrian D Hobson
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester Massachusetts 01605 USA
| |
Collapse
|
21
|
van Haaren C, Byrne B, Kazarian SG. Study of Monoclonal Antibody Aggregation at the Air-Liquid Interface under Flow by ATR-FTIR Spectroscopic Imaging. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:5858-5868. [PMID: 38445553 PMCID: PMC10956494 DOI: 10.1021/acs.langmuir.3c03730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024]
Abstract
Throughout bioprocessing, transportation, and storage, therapeutic monoclonal antibodies (mAbs) experience stress conditions that may cause protein unfolding and/or chemical modifications. Such structural changes may lead to the formation of aggregates, which reduce mAb potency and may cause harmful immunogenic responses in patients. Therefore, aggregates need to be detected and removed or ideally prevented from forming. Air-liquid interfaces, which arise during various stages of bioprocessing, are one of the stress factors causing mAb aggregation. In this study, the behavior of an immunoglobulin G (IgG) at the air-liquid interface was investigated under flow using macro attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopic imaging. This chemically specific imaging technique allows observation of adsorption of IgG to the air-liquid interface and detection of associated secondary structural changes. Chemical images revealed that IgG rapidly accumulated around an injected air bubble under flow at 45 °C; however, no such increase was observed at 25 °C. Analysis of the second derivative spectra of IgG at the air-liquid interface revealed changes in the protein secondary structure associated with increased intermolecular β-sheet content, indicative of aggregated IgG. The addition of 0.01% w/v polysorbate 80 (PS80) reduced the amount of IgG at the air-liquid interface in a static setup at 30 °C; however, this protective effect was lost at 45 °C. These results suggest that the presence of air-liquid interfaces under flow may be detrimental to mAb stability at elevated temperatures and demonstrate the power of ATR-FTIR spectroscopic imaging for studying the structural integrity of mAbs under bioprocessing conditions.
Collapse
Affiliation(s)
- Céline van Haaren
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
| | - Bernadette Byrne
- Department
of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, U.K.
| | - Sergei G. Kazarian
- Department
of Chemical Engineering, Imperial College
London, South Kensington Campus, London SW7 2AZ, U.K.
| |
Collapse
|
22
|
Yoshikawa M, Senda M, Nakamura H, Oda-Ueda N, Ueda T, Senda T, Ohkuri T. Stabilization of adalimumab Fab through the introduction of disulfide bonds between the variable and constant domains. Biochem Biophys Res Commun 2024; 700:149592. [PMID: 38295648 DOI: 10.1016/j.bbrc.2024.149592] [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: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/17/2024]
Abstract
Fab is a promising format for antibody drug. Therefore, efforts have been made to improve its thermal stability for therapeutic and commercial use. So far, we have attempted to introduce a disulfide bond into the Fab fragment to improve its thermal stability and demonstrated that it is possible to do this without sacrificing its biochemical function. In this study, to develop a novel stabilization strategy for Fab, we attempted to introduce a disulfide bond between the variable and constant domains and prepared three variants of Fab; H:G10C + H:P210C, L:P40C + L:E165C, and H:G10C + H:P210C + L:P40C + L:E165C. Differential scanning calorimetry measurements showed that each of these variants had improved thermal stability. In addition, the variants with two disulfide bonds demonstrated a 6.5 °C increase in their denaturation temperatures compared to wild-type Fab. The introduction of disulfide bonds was confirmed by X-ray crystallography, and the variants retained their antigen-binding activity. The variants were also found to be less aggregative than the wild type. Our results demonstrate that the introduction of a disulfide bond between the variable and constant domains significantly improves the thermal stability of Fab.
Collapse
Affiliation(s)
| | - Miki Senda
- Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Japan
| | | | - Naoko Oda-Ueda
- Faculty of Pharmaceutical Sciences, Sojo University, Japan
| | - Tadashi Ueda
- Graduate School of Pharmaceutical Sciences, Kyushu University, Japan
| | - Toshiya Senda
- Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Japan; Department of Accelerator Science, School of High Energy Accelerator Science, SOKENDAI, Japan; Faculty of Pure and Applied Sciences, University of Tsukuba, Japan
| | | |
Collapse
|
23
|
Brunzell E, Sigfridsson K, Gedda L, Edwards K, Bergström LM. Investigation of supramolecular structures in various aqueous solutions of an amyloid forming peptide using small-angle X-ray scattering. SOFT MATTER 2024; 20:2272-2279. [PMID: 38353286 DOI: 10.1039/d3sm01172k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Aggregation of peptide molecules into amyloid fibrils is a characteristic feature of several degenerative diseases. However, the details behind amyloid-formation, and other self-assembled peptide aggregates, remain poorly understood. In this study, we have used small-angle X-ray scattering (SAXS), static and dynamic light scattering (SLS and DLS) as well as cryogenic transmission electron microscopy (cryo-TEM) to determine the structural geometry of self-assembled peptide aggregates in various dilute aqueous solutions. Pramlintide was used as a model peptide to assess the aggregation behaviour of an amyloid-forming peptide. The effects of adding sodium chloride (NaCl), sodium thiocyanate (NaSCN), and sodium fluoride (NaF) and the co-solvent dimethyl sulfoxide (DMSO) on the aggregation behaviour were studied. Our scattering data analysis demonstrates that small oligomeric fibrils aggregate to form networks of supramolecular assemblies with fractal dimensions. The choice of anion in small amounts of added salt has a significant impact on the size of the fibrils as well as on the fractal dimensions of supramolecular clusters. In DMSO the fractal dimension decreased with increasing DMSO concentration, indicating the formation of a less compact structure of the supramolecular assemblies.
Collapse
Affiliation(s)
- Ellen Brunzell
- Department of Medicinal Chemistry, Pharmaceutical Physical Chemistry, Uppsala University, Uppsala 751 23, Sweden.
| | - Kalle Sigfridsson
- Advanced Drug Delivery, Pharmaceutical Science, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Lars Gedda
- Department of Chemistry-Ångström, Uppsala University, P.O. Box 573, Uppsala 751 23, Sweden
| | - Katarina Edwards
- Department of Chemistry-Ångström, Uppsala University, P.O. Box 573, Uppsala 751 23, Sweden
| | - L Magnus Bergström
- Department of Medicinal Chemistry, Pharmaceutical Physical Chemistry, Uppsala University, Uppsala 751 23, Sweden.
| |
Collapse
|
24
|
Escobar ELN, Vaclaw MC, Lozenski JT, Dhar P. Using Passive Microrheology to Measure the Evolution of the Rheological Properties of NIST mAb Formulations during Adsorption to the Air-Water Interface. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:4789-4800. [PMID: 38379175 DOI: 10.1021/acs.langmuir.3c03658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
The development of novel protein-based therapeutics, such as monoclonal antibodies (mAbs), is often limited due to challenges associated with maintaining the stability of these formulations during manufacturing, storage, and clinical administration. An undesirable consequence of the instability of protein therapeutics is the formation of protein particles. MAbs can adsorb to interfaces and have the potential to undergo partial unfolding as well as to form viscoelastic gels. Further, the viscoelastic properties may be correlated with their aggregation potential. In this work, a passive microrheology technique was used to correlate the evolution of surface adsorption with the evolution of surface rheology of the National Institute of Standards and Technology (NIST) mAb reference material (NIST mAb) and interface-induced subvisible protein particle formation. The evolution of the surface adsorption and interfacial shear rheological properties of the NIST mAb was recorded in four formulation conditions: two different buffers (histidine vs phosphate-buffered saline) and two different pHs (6.0 and 7.6). Our results together demonstrate the existence of multiple stages for both surface adsorption and surface rheology, characterized by an induction period that appears to be purely viscous, followed by a sharp increase in protein molecules at the interface when the film rheology is viscoelastic and ultimately a slowdown in the surface adsorption that corresponds to the formation of solid-like or glassy films at the interface. When the transitions between the different stages occurred, they were dependent on the buffer/pH of the formulations. The onset of these transitions can also be correlated to the number of protein particles formed at the interface. Finally, the addition of polysorbate 80, an FDA-approved surfactant used to mitigate protein particle formation, led to the interface being surfactant-dominated, and the resulting interface remained purely viscous.
Collapse
Affiliation(s)
- Estephanie Laura Nottar Escobar
- Department of Chemical and Petroleum Engineering, The University of Kansas, 1530W 15th Street, Lawrence, Kansas 66045, United States
| | - M Coleman Vaclaw
- Bioengineering Program, School of Engineering, The University of Kansas, 1530W 15th Street, Lawrence, Kansas 66045, United States
| | - Joseph T Lozenski
- Department of Chemical and Petroleum Engineering, The University of Kansas, 1530W 15th Street, Lawrence, Kansas 66045, United States
| | - Prajnaparamita Dhar
- Department of Chemical and Petroleum Engineering, The University of Kansas, 1530W 15th Street, Lawrence, Kansas 66045, United States
| |
Collapse
|
25
|
Escobar ELN, Griffin VP, Dhar P. Correlating Surface Activity with Interface-Induced Aggregation in a High-Concentration mAb Solution. Mol Pharm 2024; 21:1490-1500. [PMID: 38385557 DOI: 10.1021/acs.molpharmaceut.3c01125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Interface-induced aggregation resulting in protein particle formation is an issue during the manufacturing and storage of protein-based therapeutics. High-concentration formulations of therapeutic proteins are even more prone to protein particle formation due to increased protein-protein interactions. However, the dependence of interface-induced protein particle formation on bulk protein concentration is not understood. Furthermore, the formation of protein particles is often mitigated by the addition of polysorbate-based surfactants. However, the details of surfactant-protein interactions that prevent protein particle formation at high concentrations remain unclear. In this work, a tensiometer technique was used to evaluate the surface pressure of an industrially relevant mAb at different bulk concentrations, and in the absence and presence of a polysorbate-based surfactant, polysorbate 20 (PS20). The adsorption kinetics was correlated with subvisible protein particle formation at the air-water interface and in the bulk protein solution using a microflow imaging technique. Our results showed that, in the absence of any surfactant, the number of subvisible particles in the bulk protein solutions increased linearly with mAb concentration, while the number of protein particles measured at the interface showed a logarithmic dependence on bulk protein concentration. In the presence of surfactants above the critical micelle concentration (CMC), our results for low-concentration mAb solutions (10 mg/mL) showed an interface that is surfactant-dominated, and particle characterization results showed that the addition of the surfactant led to reduced particle formation. In contrast, for the highest concentration (170 mg/mL), coadsorption of proteins and surfactants was observed at the air-water interface, even for surfactant formulations above CMC and the surfactant did not mitigate subvisible particle formation. Our results taken together provide evidence that the ratio between the surfactant and mAb molecules is an important consideration when formulating high-concentration mAb therapeutics to prevent unwanted aggregation.
Collapse
Affiliation(s)
- Estephanie L N Escobar
- Department of Chemical and Petroleum Engineering, The University of Kansas, 1530W 15th Street, Lawrence, Kansas 66045, United States
| | - Valerie P Griffin
- Department of Chemical and Petroleum Engineering, The University of Kansas, 1530W 15th Street, Lawrence, Kansas 66045, United States
| | - Prajnaparamita Dhar
- Department of Chemical and Petroleum Engineering, The University of Kansas, 1530W 15th Street, Lawrence, Kansas 66045, United States
| |
Collapse
|
26
|
Cernosek T, Jain N, Dalphin M, Behrens S, Wunderli P. Accelerated development of a SEC-HPLC procedure for purity analysis of monoclonal antibodies using design of experiments. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1235:124037. [PMID: 38335765 DOI: 10.1016/j.jchromb.2024.124037] [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: 12/05/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024]
Abstract
The complex structure of biopharmaceutical products poses an inherent need for their thorough characterization to ensure product quality, safety, and efficacy. Analytical size exclusion chromatography (SEC) is a widely used technique throughout the development and manufacturing of monoclonal antibodies (mAbs) which quantifies product size variants such as aggregates and fragments. Aggregate and fragment content are critical quality attributes (CQAs) in mAb products, as higher contents of such size heterogeneities impact product quality. Historically, SEC methods have achieved sufficient separation between the high molecular weight (HMW) species and the main product. In contrast, some low molecular weight (LMW) species are often not sufficiently different in molecular mass from the main product, making it difficult to achieve appropriate resolutions between the two species. This lack of resolution makes it difficult to consistently quantify the LMW species in mAb-based therapeutics. The following work uses a design of experiments (DoE) approach to establish a robust analytical SEC procedure by evaluating SEC column types and mobile phase compositions using two mAb products with different physiochemical properties. The resulting optimized procedure using a Waters™ BioResolve column exhibits an improved ability to resolve and quantify mAb size variants, highlighting improvement in the resolution of the LMW species. Additionally, the addition of L-arginine as a mobile phase additive showed to reduce secondary interactions and was beneficial in increasing the recoveries of the HMW species.
Collapse
Affiliation(s)
- Terezie Cernosek
- Catalent Biologics, Madison, WI, USA; Keck Graduate Institute of Applied Life Sciences, Claremont, CA, USA.
| | | | | | - Sue Behrens
- Keck Graduate Institute of Applied Life Sciences, Claremont, CA, USA
| | | |
Collapse
|
27
|
Harris CT, Cohen S. Reducing Immunogenicity by Design: Approaches to Minimize Immunogenicity of Monoclonal Antibodies. BioDrugs 2024; 38:205-226. [PMID: 38261155 PMCID: PMC10912315 DOI: 10.1007/s40259-023-00641-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 01/24/2024]
Abstract
Monoclonal antibodies (mAbs) have transformed therapeutic strategies for various diseases. Their high specificity to target antigens makes them ideal therapeutic agents for certain diseases. However, a challenge to their application in clinical practice is their potential risk to induce unwanted immune response, termed immunogenicity. This challenge drives the continued efforts to deimmunize these protein therapeutics while maintaining their pharmacokinetic properties and therapeutic efficacy. Because mAbs hold a central position in therapeutic strategies against an array of diseases, the importance of conducting comprehensive immunogenicity risk assessment during the drug development process cannot be overstated. Such assessment necessitates the employment of in silico, in vitro, and in vivo strategies to evaluate the immunogenicity risk of mAbs. Understanding the intricacies of the mechanisms that drive mAb immunogenicity is crucial to improving their therapeutic efficacy and safety and developing the most effective strategies to determine and mitigate their immunogenic risk. This review highlights recent advances in immunogenicity prediction strategies, with a focus on protein engineering strategies used throughout development to reduce immunogenicity.
Collapse
Affiliation(s)
- Chantal T Harris
- Department of BioAnalytical Sciences, Genentech Inc., South San Francisco, CA, 94080-4990, USA
| | - Sivan Cohen
- Department of BioAnalytical Sciences, Genentech Inc., South San Francisco, CA, 94080-4990, USA.
| |
Collapse
|
28
|
Haegens LL, Huiskes VJB, Bekker CL, van den Bemt BJF. Effect of a smart temperature logger on correctly storing biological disease-modifying antirheumatic drugs at home: a pre-post study. Eur J Hosp Pharm 2024:ejhpharm-2023-004028. [PMID: 38383141 DOI: 10.1136/ejhpharm-2023-004028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024] Open
Abstract
OBJECTIVES Biological disease-modifying antirheumatic drugs (bDMARDs) require specific storage temperatures, but are frequently stored outside the recommended range of 2-8°C. As incorrect storage may affect therapy effectiveness and consequently lead to higher disease activity, compliance with recommended storage temperatures should be improved. eHealth interventions can provide insight into storage temperatures and alerts in case of deviations from recommended temperatures. Therefore, this study aims to assess the effect of a smart temperature logger on correctly storing bDMARDs at home by patients with rheumatic diseases. METHODS A pre-post study was performed in a hospital in the Netherlands. The baseline period consisted of 12 weeks of storage temperature measurement with a passive temperature logger, and the intervention period consisted of 12 weeks of storage temperature measurement with a smart temperature logger. This smart logger included a smartphone application which provided insight into storage temperatures and real-time alerts when exceeding recommended temperatures. The main outcome measure was the difference in the number of patients who stored their bDMARDs correctly between baseline and intervention. Secondary outcomes were the difference in the proportion of measurement time within 2-8°C between baseline and intervention, the distribution of measurement time among temperature categories, and the patient's acceptance measured using a questionnaire based on the Technology Acceptance Model. RESULTS In total, 48 participants (median age 55 years (IQR 47-64), 53% male) were analysed. The proportion of participants correctly storing bDMARDs increased from 18.8% (n=9) during baseline to 39.6% (n=19) during intervention (p=0.004). The median proportion of measurement time between 2-8°C improved by 6% (IQR 0-34%) (p<0.0001). Technology acceptance was scored as moderate. CONCLUSIONS Temperature monitoring and real-time feedback with a smart temperature logger shows potential to improve at-home storage of bDMARDs, provided that continuous connection is realised to ensure real-time alerts and data collection.
Collapse
Affiliation(s)
- Lex L Haegens
- Department of Research, Sint Maartenskliniek, Nijmegen, The Netherlands
| | - Victor J B Huiskes
- Department of Pharmacy, Sint Maartenskliniek, Nijmegen, Gelderland, The Netherlands
- Department of Pharmacy, Radboudumc, Nijmegen, Gelderland, The Netherlands
| | - Charlotte L Bekker
- Department of Pharmacy, Radboudumc, Nijmegen, Gelderland, The Netherlands
| | - Bart J F van den Bemt
- Department of Pharmacy, Sint Maartenskliniek, Nijmegen, Gelderland, The Netherlands
- Department of Pharmacy, Radboudumc, Nijmegen, Gelderland, The Netherlands
| |
Collapse
|
29
|
Floyd JA, Gillespie AJ, Nightlinger NS, Siska C, Kerwin BA. The Development of a Novel Aflibercept Formulation for Ocular Delivery. J Pharm Sci 2024; 113:366-376. [PMID: 38042344 DOI: 10.1016/j.xphs.2023.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Aflibercept is a recombinant fusion protein that is commercially available for several ocular diseases impacting millions of people worldwide. Here, we use a case study approach to examine alternative liquid formulations for aflibercept for ocular delivery, utilizing different stabilizers, buffering agents, and surfactants with the goal of improving the thermostability to allow for limited storage outside the cold chain. The formulations were developed by studying the effects of pH changes, substituting amino acids for sucrose and salt, and using polysorbate 80 or poloxamer 188 instead of polysorbate 20. A formulation containing acetate, proline, and poloxamer 188 had lower rates of aggregate formation at 4, 30, and 40°C when compared to the marketed commercial formulation containing phosphate, sucrose, sodium chloride, and polysorbate 20. Further studies examining subvisible particles after exposure to a transport stress and long-term stability at 4°C, post-translational modifications by multi-attribute method, purity by reduced and non-reduced capillary electrophoresis, and potency by cell proliferation also demonstrated a comparable or improved stability for the enhanced formulation of acetate, proline, and poloxamer 188. This enhanced stability could enable limited storage outside of the cold chain, allowing for easier distribution in low to middle income countries.
Collapse
Affiliation(s)
- J Alaina Floyd
- Just- Evotec Biologics, 401 Terry Ave N., Seattle, WA 98109, USA.
| | | | | | - Christine Siska
- Just- Evotec Biologics, 401 Terry Ave N., Seattle, WA 98109, USA
| | - Bruce A Kerwin
- Just- Evotec Biologics, 401 Terry Ave N., Seattle, WA 98109, USA.
| |
Collapse
|
30
|
Chen YL, Lee J, Liu Z, Strych U, Bottazzi ME, Lin YP, Chen WH. Biophysical and biochemical characterization of a recombinant Lyme disease vaccine antigen, CspZ-YA. Int J Biol Macromol 2024; 259:129295. [PMID: 38211914 DOI: 10.1016/j.ijbiomac.2024.129295] [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: 10/19/2023] [Revised: 12/19/2023] [Accepted: 01/05/2024] [Indexed: 01/13/2024]
Abstract
Lyme disease, caused by Lyme Borrelia spirochetes, is the most common vector-borne illness in the United States. Despite its global significance, with an estimated 14.5 % seroprevalence, there is currently no licensed vaccine. Previously, we demonstrated that CspZ-YA protein conferred protection against Lyme Borrelia infection, making it a promising vaccine candidate. However, such a protein was tagged with hexahistidine, and thus not preferred for vaccine development; furthermore, the formulation to stabilize the protein was understudied. In this work, we developed a two-step purification process for tag-free E. coli-expressed recombinant CspZ-YA. We further utilized various bioassays to analyze the protein and determine the suitable buffer system for long-term storage and formulation as a vaccine immunogen. The results indicated that a buffer with a pH between 6.5 and 8.5 stabilized CspZ-YA by reducing its surface hydrophobicity and colloidal interactions. Additionally, low pH values induced a change in local spatial conformation and resulted in a decrease in α-helix content. Lastly, an optimal salinity of 22-400 mM at pH 7.5 was found to be important for its stability. Collectively, this study provides a fundamental biochemical and biophysical understanding and insights into the ideal stabilizing conditions to produce CspZ-YA recombinant protein for use in vaccine formulation and development.
Collapse
Affiliation(s)
- Yi-Lin Chen
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Jungsoon Lee
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Zhuyun Liu
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Ulrich Strych
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Maria Elena Bottazzi
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Biology, Baylor University, Waco, TX, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA; Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA.
| | - Wen-Hsiang Chen
- Department of Pediatrics, Division of Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA.
| |
Collapse
|
31
|
Kaygisiz K, Rauch‐Wirth L, Iscen A, Hartenfels J, Kremer K, Münch J, Synatschke CV, Weil T. Peptide Amphiphiles as Biodegradable Adjuvants for Efficient Retroviral Gene Delivery. Adv Healthc Mater 2024; 13:e2301364. [PMID: 37947246 PMCID: PMC11468294 DOI: 10.1002/adhm.202301364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/20/2023] [Indexed: 11/12/2023]
Abstract
Retroviral gene delivery is the key technique for in vitro and ex vivo gene therapy. However, inefficient virion-cell attachment resulting in low gene transduction efficacy remains a major challenge in clinical applications. Adjuvants for ex vivo therapy settings need to increase transduction efficiency while being easily removed or degraded post-transduction to prevent the risk of venous embolism after infusing the transduced cells back to the bloodstream of patients, yet no such peptide system have been reported thus far. In this study, peptide amphiphiles (PAs) with a hydrophobic fatty acid and a hydrophilic peptide moiety that reveal enhanced viral transduction efficiency are introduced. The PAs form β-sheet-rich fibrils that assemble into positively charged aggregates, promoting virus adhesion to the cell membrane. The block-type amphiphilic sequence arrangement in the PAs ensures efficient cell-virus interaction and biodegradability. Good biodegradability is observed for fibrils forming small aggregates and it is shown that via molecular dynamics simulations, the fibril-fibril interactions of PAs are governed by fibril surface hydrophobicity. These findings establish PAs as additives in retroviral gene transfer, rivalling commercially available transduction enhancers in efficiency and degradability with promising translational options in clinical gene therapy applications.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Lena Rauch‐Wirth
- Institute of Molecular VirologyUlm University Medical CenterMeyerhofstraße 189081UlmGermany
| | - Aysenur Iscen
- Polymer Theory DepartmentMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Jan Hartenfels
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Kurt Kremer
- Polymer Theory DepartmentMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Jan Münch
- Institute of Molecular VirologyUlm University Medical CenterMeyerhofstraße 189081UlmGermany
| | - Christopher V. Synatschke
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Tanja Weil
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| |
Collapse
|
32
|
Fast J, Christian T, Crul M, Jiskoot W, Nejadnik MR, Medina A, Radwick A, Sreedhara A, Tole H. Use of Closed System Transfer Devices (CSTDs) with Protein-Based Therapeutic Drugs-A Non-Solution for a Non-Problem? J Pharm Sci 2024; 113:298-305. [PMID: 37984700 DOI: 10.1016/j.xphs.2023.11.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Affiliation(s)
- Jonas Fast
- Pharmaceutical Development, F. Hoffmann-La Roche Ltd. CH-4070 Basel, Switzerland.
| | | | - Mirjam Crul
- Amsterdam University Medical Center, Department of Clinical Pharmacology and Pharmacy, Amsterdam, the Netherlands
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden University, the Netherlands; Coriolis Pharma, Martinsried, Germany
| | - M Reza Nejadnik
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Annette Medina
- Dosage Form Design and Development, AstraZeneca, Gaithersburg, MD, USA
| | | | | | - Hugh Tole
- Occupational Health, Hygiene &Toxicology, Genentech Inc., Little Falls, NJ, USA
| |
Collapse
|
33
|
Moino C, Artusio F, Pisano R. Shear stress as a driver of degradation for protein-based therapeutics: More accomplice than culprit. Int J Pharm 2024; 650:123679. [PMID: 38065348 DOI: 10.1016/j.ijpharm.2023.123679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/08/2024]
Abstract
Protein degradation is a major concern for protein-based therapeutics. It may alter the biological activity of the product and raise the potential for undesirable effects on the patients. Among the numerous drivers of protein degradation, shear stress has been the focus around which much work has revolved since the 1970s. In the pharmaceutical realm, the product is often processed through several unit operations, which include mixing, pumping, filtration, filling, and atomization. Nonetheless, the drug might be exposed to significant shear stresses, which might cooperatively contribute to product degradation, together with interfacial stress. This review presents fundamentals of shear stress about protein structure, followed by an overview of the drivers of product degradation. The impact of shear stress on protein stability in different unit operations is then presented, and recommendations for limiting the adverse effects on the biopharmaceutical formulations are outlined. Finally, several devices used to explore the effects of shear stress are discussed.
Collapse
Affiliation(s)
- Camilla Moino
- Department of Applied Science and Technology, Politecnico di Torino, 24 Corso Duca degli Abruzzi, Torino 10129, Italy
| | - Fiora Artusio
- Department of Applied Science and Technology, Politecnico di Torino, 24 Corso Duca degli Abruzzi, Torino 10129, Italy
| | - Roberto Pisano
- Department of Applied Science and Technology, Politecnico di Torino, 24 Corso Duca degli Abruzzi, Torino 10129, Italy.
| |
Collapse
|
34
|
Cucuzza S, Brosig S, Serno T, Bechtold-Peters K, Cerar J, Kammüller M, Gallou F. Modular and tunable alternative surfactants for biopharmaceuticals provide insights into Surfactant's Structure-Function relationship. Int J Pharm 2024; 650:123692. [PMID: 38081561 DOI: 10.1016/j.ijpharm.2023.123692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Surface-induced aggregation of protein therapeutics is opposed by employing surfactants, which are ubiquitously used in drug product development, with polysorbates being the gold standard. Since poloxamer 188 is currently the only generally accepted polysorbate alternative, but cannot be ubiquitously applied, there is a strong need to develop surfactant alternatives for protein biologics that would complement and possibly overcome known drawbacks of existing surfactants. Yet, a severe lack of structure-function relationship knowledge complicates the development of new surfactants. Herein, we perform a systematic analysis of the structure-function relationship of three classes of novel alternative surfactants. Firstly, the mode of action is thoroughly characterized through tensiometry, calorimetry and MD simulations. Secondly, the safety profiles are evaluated through cell-based in vitro assays. Ultimately, we could conclude that the alternative surfactants investigated possess a mode of action and safety profile comparable to polysorbates. Moreover, the biophysical patterns elucidated here can be exploited to precisely tune the features of future surfactant designs.
Collapse
Affiliation(s)
- Stefano Cucuzza
- Novartis Pharma AG, TRD Biologics & CGT, GDD, 4002 Basel, Switzerland
| | - Sebastian Brosig
- Novartis Pharma AG, TRD Biologics & CGT, GDD, 4002 Basel, Switzerland
| | - Tim Serno
- Novartis Pharma AG, TRD Biologics & CGT, GDD, 4002 Basel, Switzerland
| | | | - Jure Cerar
- Novartis Pharma AG, TRD Biologics & CGT, GDD, 1234 Menges, Slovenia
| | | | | |
Collapse
|
35
|
King TE, Humphrey JR, Laughton CA, Thomas NR, Hirst JD. Optimizing Excipient Properties to Prevent Aggregation in Biopharmaceutical Formulations. J Chem Inf Model 2024; 64:265-275. [PMID: 38113509 PMCID: PMC10777730 DOI: 10.1021/acs.jcim.3c01898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Excipients are included within protein biotherapeutic solution formulations to improve colloidal and conformational stability but are generally not designed for the specific purpose of preventing aggregation and improving cryoprotection in solution. In this work, we have explored the relationship between the structure and antiaggregation activity of excipients by utilizing coarse-grained molecular dynamics modeling of protein-excipient interaction. We have studied human serum albumin as a model protein, and we report the interaction of 41 excipients (polysorbates, fatty alcohol ethoxylates, fatty acid ethoxylates, phospholipids, glucosides, amino acids, and others) in terms of the reduction of solvent accessible surface area of aggregation-prone regions, proposed as a mechanism of aggregation prevention. Polyoxyethylene sorbitan had the greatest degree of interaction with aggregation-prone regions, decreasing the solvent accessible surface area of APRs by 20.7 nm2 (40.1%). Physicochemical descriptors generated by Mordred are employed to probe the structure-property relationship using partial least-squares regression. A leave-one-out cross-validated model had a root-mean-square error of prediction of 4.1 nm2 and a mean relative error of prediction of 0.077. Generally, longer molecules with a large number of alcohol-terminated PEG units tended to interact more, with qualitatively different protein interactions, wrapping around the protein. Shorter or less ethoxylated compounds tend to form hemimicellar clusters at the protein surface. We propose that an improved design would feature many short chains of 5 to 10 PEG units in many distinct branches and at least some hydrophobic content in the form of medium-length or greater aliphatic chains (i.e., six or more carbon atoms). The combination of molecular dynamics simulation and quantitative modeling is an important first step in an all-purpose protein-independent model for the computer-aided design of stabilizing excipients.
Collapse
Affiliation(s)
- Toby E. King
- Biodiscovery
Institute, School of Pharmacy, University Park, Nottingham NG7 2RD, U.K.
| | | | - Charles A. Laughton
- Biodiscovery
Institute, School of Pharmacy, University Park, Nottingham NG7 2RD, U.K.
| | - Neil R. Thomas
- Biodiscovery
Institute, School of Chemistry, University Park, Nottingham NG7 2RD, U.K.
| | | |
Collapse
|
36
|
Li Z, Du X, Wang YMC. A survey of FDA Approved Monoclonal Antibodies and Fc-fusion Proteins for Manufacturing Changes and Comparability Assessment. Pharm Res 2024; 41:13-27. [PMID: 37910341 DOI: 10.1007/s11095-023-03627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE Manufacturing changes occur commonly throughout stages of biologics development and may result in product quality attribute changes. As changes in critical quality attributes have the potential to affect clinical safety and efficacy of products, it is imperative to ensure the quality and clinical performance before introducing the after-change products. Thus, we embarked on this project to understand what data have supported the manufacturing changes for licensed products with pre- and post-approval changes. METHODS We surveyed the manufacturing changes of 85 monoclonal antibodies and 10 Fc fusion proteins approved by the Food and Drug Administration as of December 25, 2021. After collecting the type and timing of changes for these products, we investigated the approaches that provided supporting data for the changes. The source documents included reports submitted by applicants and FDA's regulatory reviews. RESULTS Analytical comparability was assessed to support all identified manufacturing changes. Supporting clinical data were available in 92% of these manufacturing changes; including data from pharmacokinetic comparability studies alone (3%), other studies on efficacy or safety (70%) and a combination of both (19%). Clinical pharmacokinetic comparability data contributed to supporting substantial changes, such as host cell type or master cell bank changes, concentration or formulation changes, and changes from pre-filled syringes to autoinjectors, especially when introduced after completing pivotal studies. CONCLUSION Our comprehensive retrospective analysis provides an understanding of the regulatory experience and industry practice, which could facilitate developing appropriate comparability approaches to support manufacturing changes in the future.
Collapse
Affiliation(s)
- Zhe Li
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (OCP/CDER/FDA), 10903 New Hampshire Avenue, Silver Spring, MD, USA.
| | - Xiulian Du
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (OCP/CDER/FDA), 10903 New Hampshire Avenue, Silver Spring, MD, USA
| | - Yow-Ming C Wang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (OCP/CDER/FDA), 10903 New Hampshire Avenue, Silver Spring, MD, USA
| |
Collapse
|
37
|
Chowdhury S, Sarkar N. Exploring the potential of amyloids in biomedical applications: A review. Biotechnol Bioeng 2024; 121:26-38. [PMID: 37822225 DOI: 10.1002/bit.28569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/31/2023] [Accepted: 09/24/2023] [Indexed: 10/13/2023]
Abstract
Amyloid is defined as a fibrous quaternary structure formed by assembling protein or peptide monomers into intermolecularly hydrogen linked β-sheets. There is a prevalent issue with protein aggregation and the buildup of amyloid molecules, which results in human neurological illnesses including Alzheimer's and Parkinson's. But it is now evident that many organisms, like bacteria, fungi as well as humans, use the same fibrillar structure to carry out a variety of biological functions, such as structure and protection supporting interface transitions and cell-cell recognition, protein control and storage, epigenetic inheritance, and memory. Recent discoveries of self-assembling amyloidogenic peptides and proteins, based on the amyloid core structure, give rise to interesting biomaterials with potential uses in numerous industries. These functions dramatically diverge from the initial conception of amyloid fibrils as intrinsically diseased entities. Apart from the natural ability of amyloids to spontaneously arrange themselves and their exceptional material characteristics, this aspect has prompted extensive research into engineering artificial amyloids for generating various nanostructures, molecular substances, and combined materials. Here, we discuss significant developments in the artificial design of useful amyloids as well as how amyloid materials serve as examples of how function emerges from protein self-assembly at various length scales.
Collapse
Affiliation(s)
- Srijita Chowdhury
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Nandini Sarkar
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela, Odisha, India
| |
Collapse
|
38
|
Tsai WTK, Li Y, Yin Z, Tran P, Phung Q, Zhou Z, Peng K, Qin D, Tam S, Spiess C, Brumm J, Wong M, Ye Z, Wu P, Cohen S, Carter PJ. Nonclinical immunogenicity risk assessment for knobs-into-holes bispecific IgG 1 antibodies. MAbs 2024; 16:2362789. [PMID: 38845069 PMCID: PMC11164226 DOI: 10.1080/19420862.2024.2362789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024] Open
Abstract
Bispecific antibodies, including bispecific IgG, are emerging as an important new class of antibody therapeutics. As a result, we, as well as others, have developed engineering strategies designed to facilitate the efficient production of bispecific IgG for clinical development. For example, we have extensively used knobs-into-holes (KIH) mutations to facilitate the heterodimerization of antibody heavy chains and more recently Fab mutations to promote cognate heavy/light chain pairing for efficient in vivo assembly of bispecific IgG in single host cells. A panel of related monospecific and bispecific IgG1 antibodies was constructed and assessed for immunogenicity risk by comparison with benchmark antibodies with known low (Avastin and Herceptin) or high (bococizumab and ATR-107) clinical incidence of anti-drug antibodies. Assay methods used include dendritic cell internalization, T cell proliferation, and T cell epitope identification by in silico prediction and MHC-associated peptide proteomics. Data from each method were considered independently and then together for an overall integrated immunogenicity risk assessment. In toto, these data suggest that the KIH mutations and in vitro assembly of half antibodies do not represent a major risk for immunogenicity of bispecific IgG1, nor do the Fab mutations used for efficient in vivo assembly of bispecifics in single host cells. Comparable or slightly higher immunogenicity risk assessment data were obtained for research-grade preparations of trastuzumab and bevacizumab versus Herceptin and Avastin, respectively. These data provide experimental support for the common practice of using research-grade preparations of IgG1 as surrogates for immunogenicity risk assessment of their corresponding pharmaceutical counterparts.
Collapse
Affiliation(s)
- Wen-Ting K. Tsai
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA, USA
| | - Yinyin Li
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc, South San Francisco, CA, USA
| | - Zhaojun Yin
- Department of Bioanalytical Sciences, Genentech, Inc, South San Francisco, CA, USA
| | - Peter Tran
- Department of Bioanalytical Sciences, Genentech, Inc, South San Francisco, CA, USA
| | - Qui Phung
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc, South San Francisco, CA, USA
| | - Zhenru Zhou
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc, South San Francisco, CA, USA
| | - Kun Peng
- Department of Bioanalytical Sciences, Genentech, Inc, South San Francisco, CA, USA
| | - Dan Qin
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc, South San Francisco, CA, USA
| | - Sien Tam
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc, South San Francisco, CA, USA
| | - Christoph Spiess
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA, USA
| | - Jochen Brumm
- Department of Nonclinical Biostatistics, Genentech, Inc, South San Francisco, CA, USA
| | - Manda Wong
- Department of Structural Biology, Genentech, Inc, South San Francisco, CA, USA
| | - Zhengmao Ye
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc, South San Francisco, CA, USA
| | - Patrick Wu
- Department of Bioanalytical Sciences, Genentech, Inc, South San Francisco, CA, USA
| | - Sivan Cohen
- Department of Bioanalytical Sciences, Genentech, Inc, South San Francisco, CA, USA
| | - Paul J. Carter
- Department of Antibody Engineering, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
39
|
Oyama K, Nakakido M, Ohkuri T, Nakamura H, Tsumoto K, Ueda T. Enhancing thermal stability in the CH 2 domain to suppress aggregation through the introduction of simultaneous disulfide bonds in Pichia pastoris. Protein Sci 2023; 32:e4831. [PMID: 37924310 PMCID: PMC10680342 DOI: 10.1002/pro.4831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/21/2023] [Accepted: 11/01/2023] [Indexed: 11/06/2023]
Abstract
Protein aggregations decrease production yields and impair the efficacy of therapeutics. The CH2 domain is a crucial part of the constant region of human IgG. But, it is also the least stable domain in IgG, which can result in antibody instability and aggregation problems. We created a novel mutant of the CH2 domain (T250C/L314C, mut10) by introducing a disulfide bond and expressed it using Pichia pastoris. The mut10 variant exhibited enhanced thermal stability, resistance to enzymatic degradation, and reduced aggregation in comparison to the original CH2 domain. However, it was less stable than mut20 (L242C/K334C), which is the variant prepared in a previous study (Gong et al., J. Biol. Chem., 2009). A more advanced mutant, mut25, was created by combining mut10 and mut20. Mut25 artificially contains two disulfide bonds. The new mutant, mut25, showed enhanced thermal stability, increased resistance to enzymatic digestion, and reduced aggregation in comparison to mut20. According to our knowledge, mut25 achieves an unprecedented level of stability among the humanized whole CH2 domains that have been reported so far. Mut25 has the potential to serve as a new platform for antibody therapeutics due to its ability to reduce immunogenicity by decreasing aggregation.
Collapse
Affiliation(s)
- Kosuke Oyama
- Graduate School of Pharmaceutical SciencesKyushu UniversityFukuokaJapan
| | - Makoto Nakakido
- Graduate School of EngineeringThe University of TokyoTokyoJapan
| | | | - Hitomi Nakamura
- Faculty of Pharmaceutical SciencesSojo UniversityKumamotoJapan
| | - Kouhei Tsumoto
- Graduate School of EngineeringThe University of TokyoTokyoJapan
| | - Tadashi Ueda
- Graduate School of Pharmaceutical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
40
|
Riccardi C, Carlson DP, Graham KS, Shameem M, Kamen DE. Evaluation of the In-Use Stability of Monoclonal Antibody IV Admixtures Prepared from Drug Products Containing Polysorbate 20 Degraded by Host-Cell Lipases. J Pharm Sci 2023; 112:3045-3055. [PMID: 37643700 DOI: 10.1016/j.xphs.2023.08.020] [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: 03/10/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Host-cell lipases can be present in monoclonal antibody drug products and can degrade polysorbates present in the formulations as stabilizers. We hypothesized that the in-use stability of the IV admixture prepared from such a drug product might be impacted by decreasing levels of polysorbate 20. Host-cell lipase activity has, in fact, been observed during development of one of our therapeutic monoclonal antibody drug products. Throughout the course of the product shelf life, polysorbate 20 levels decreased but no other quality attributes of the drug product were impacted. An experimental approach was developed to simulate how the prepared IV admixture in-use stability is affected as polysorbate 20 concentration in the drug product decreased over the shelf life, and from that a minimum level of polysorbate 20 required in the drug product was determined to estimate the in-use stability of the IV admixture as the polysorbate 20 in the drug product degrades. The results indicate that although the observed degradation of polysorbate 20 does not affect quality attributes of this drug product, in-use stability of the IV admixture as a function of polysorbate degradation can be impacted and should be assessed to ensure sufficient quality.
Collapse
Affiliation(s)
- Caterina Riccardi
- Regeneron Pharmaceuticals Inc., Formulation Development, 777 Old Saw Mill River Road, Tarrytown, New York, NY 10591, USA
| | - Dane P Carlson
- Regeneron Pharmaceuticals Inc., Formulation Development, 777 Old Saw Mill River Road, Tarrytown, New York, NY 10591, USA
| | - Kenneth S Graham
- Regeneron Pharmaceuticals Inc., Formulation Development, 777 Old Saw Mill River Road, Tarrytown, New York, NY 10591, USA
| | - Mohammed Shameem
- Regeneron Pharmaceuticals Inc., Formulation Development, 777 Old Saw Mill River Road, Tarrytown, New York, NY 10591, USA
| | - Douglas E Kamen
- Regeneron Pharmaceuticals Inc., Formulation Development, 777 Old Saw Mill River Road, Tarrytown, New York, NY 10591, USA.
| |
Collapse
|
41
|
Mojtahed Poor S, Henke M, Ulshöfer T, Köhm M, Behrens F, Burkhardt H, Schiffmann S. The role of antidrug antibodies in ustekinumab therapy and the impact of methotrexate. Rheumatology (Oxford) 2023; 62:3993-3999. [PMID: 37079726 PMCID: PMC10691926 DOI: 10.1093/rheumatology/kead177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/21/2023] [Accepted: 04/02/2023] [Indexed: 04/22/2023] Open
Abstract
OBJECTIVE We investigated the impact of concomitant MTX on ustekinumab (UST) levels and antidrug antibody (ADA) formation in PsA and evaluated consequences in pharmacodynamics and pharmacokinetics. METHODS We conducted a post-hoc analysis on 112 PsA serum samples of subjects treated with open-label UST and either concomitant MTX (UST/MTX, n = 58) or placebo (UST/pbo, n = 54) obtained in a randomized (1:1), double-blind, multicentre trial. A validated antibody-binding-based multitiered testing was used to detect ADA and ADA with neutralizing capacity (nADA). The impact of MTX on UST immunogenicity was analysed by comparison of UST/pbo with UST/MTX cohorts at different time points. Patient- and disease-related predispositions for ADA formation were investigated with multiple linear regression analysis. Immunogenicity impact on pharmacokinetics, safety and efficacy was determined by cohort comparison between patients with and without ADA formation. RESULTS Over 52 weeks, 11 UST/pbo- and 19 UST/MTX-treated patients developed ADA (P > 0.05). In the UST/pbo cohort, the visit-dependent UST levels were in the range of 0.047 (0.05) -0.110 (0.07) µg/ml overall, and 0.037 (0.04)-0.091 (0.08) µg/ml in ADA-confirmed subjects. In UST/MTX-treated patients, the UST levels exhibited an intervisit variation in the range of 0.0502 (0.04)-0.106 (0.07) µg/ml overall and 0.029 (0.03)-0.097 (0.07) µg/ml in ADA positive subjects (P > 0.05). At week 52, ADA-confirmed patients did not differ significantly (P > 0.05) in safety or clinical outcomes from ADA-negative patients. CONCLUSION Concomitant MTX had no significant impact on UST immunogenicity. Furthermore, ADA formation was not associated with impairments in UST safety, efficacy or trough levels. TRIAL REGISTRATION ClinicalTrials.gov, https://clinicaltrials.gov, NCT03148860.
Collapse
Affiliation(s)
- Sorwe Mojtahed Poor
- Department of Rheumatology, Goethe-University Hospital, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Marina Henke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Thomas Ulshöfer
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Michaela Köhm
- Department of Rheumatology, Goethe-University Hospital, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Frank Behrens
- Department of Rheumatology, Goethe-University Hospital, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Harald Burkhardt
- Department of Rheumatology, Goethe-University Hospital, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Susanne Schiffmann
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| |
Collapse
|
42
|
Hada S, Burlakoti U, Kim KH, Han JS, Kim MJ, Kim NA, Jeong SH. A comprehensive evaluation of arginine and its derivatives as protein formulation stabilizers. Int J Pharm 2023; 647:123545. [PMID: 37871869 DOI: 10.1016/j.ijpharm.2023.123545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/14/2023] [Accepted: 10/21/2023] [Indexed: 10/25/2023]
Abstract
Arginine and its derivatives (such as arginine ethyl ester and acetyl arginine) have varying degrees of protein aggregation suppressor effect across different protein solutions. To understand this performance ambiguity, we evaluated the activity of arginine, acetyl arginine, and arginine ethyl ester for aggregation suppressor effect against human intravenous immunoglobulin G (IgG) solution at pH 4.8. Both arginine and its cationic derivative arginine ethyl ester in their hydrochloride salt forms significantly reduced the colloidal and conformational stability (reduced kd and Tm) of IgG. Consequently, the monomer content was decreased with an increase in subvisible particulates after agitation or thermal stress. Furthermore, compared to arginine, arginine ethyl ester with one more cationic charge and hydrochloride salt form readily precipitated IgG at temperatures higher than 25 °C. On the contrary, acetyl arginine, which mostly exists in a neutral state at pH 4.8, efficiently suppressed the formation of subvisible particles retaining a high amount of monomer owing to its higher colloidal and conformational stability. Concisely, the charged state of additives significantly impacts protein stability. This study demonstrated that contrary to popular belief, arginine and its derivatives may either enhance or suppress protein aggregation depending on their net charge and concentration.
Collapse
Affiliation(s)
- Shavron Hada
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Gyeonggi 10326, Republic of Korea.
| | - Urmila Burlakoti
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Gyeonggi 10326, Republic of Korea.
| | - Ki Hyun Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Gyeonggi 10326, Republic of Korea.
| | - Ji Soo Han
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea.
| | - Min Ji Kim
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea.
| | - Nam Ah Kim
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Muan 58554, Republic of Korea; Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Muan 58554, Republic of Korea.
| | - Seong Hoon Jeong
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Gyeonggi 10326, Republic of Korea.
| |
Collapse
|
43
|
Jha SK, Imran M, Jha LA, Hasan N, Panthi VK, Paudel KR, Almalki WH, Mohammed Y, Kesharwani P. A Comprehensive review on Pharmacokinetic Studies of Vaccines: Impact of delivery route, carrier-and its modulation on immune response. ENVIRONMENTAL RESEARCH 2023; 236:116823. [PMID: 37543130 DOI: 10.1016/j.envres.2023.116823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
The lack of knowledge about the absorption, distribution, metabolism, and excretion (ADME) of vaccines makes former biopharmaceutical optimization difficult. This was shown during the COVID-19 immunization campaign, where gradual booster doses were introduced.. Thus, understanding vaccine ADME and its effects on immunization effectiveness could result in a more logical vaccine design in terms of formulation, method of administration, and dosing regimens. Herein, we will cover the information available on vaccine pharmacokinetics, impacts of delivery routes and carriers on ADME, utilization and efficiency of nanoparticulate delivery vehicles, impact of dose level and dosing schedule on the therapeutic efficacy of vaccines, intracellular and endosomal trafficking and in vivo fate, perspective on DNA and mRNA vaccines, new generation sequencing and mathematical models to improve cancer vaccination and pharmacology, and the reported toxicological study of COVID-19 vaccines. Altogether, this review will enhance the reader's understanding of the pharmacokinetics of vaccines and methods that can be implied in delivery vehicle design to improve the absorption and distribution of immunizing agents and estimate the appropriate dose to achieve better immunogenic responses and prevent toxicities.
Collapse
Affiliation(s)
- Saurav Kumar Jha
- Department of Biomedicine, Health & Life Convergence Sciences, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea; Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India.
| | - Mohammad Imran
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Laxmi Akhileshwar Jha
- H. K. College of Pharmacy, Mumbai University, Pratiksha Nagar, Jogeshwari, West Mumbai, 400102, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Vijay Kumar Panthi
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Science, Centenary Institute and University of Technology Sydney, Sydney, 2007, Australia
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 24381, Saudi Arabia
| | - Yousuf Mohammed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
44
|
Wu KW, Chen TH, Yang TC, Wang SC, Shameem M, Graham KS. Continuous monitoring of a monoclonal antibody by size exclusion chromatography reveals a correlation between system suitability parameters and column aging. J Pharm Biomed Anal 2023; 235:115622. [PMID: 37540994 DOI: 10.1016/j.jpba.2023.115622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Size exclusion chromatography (SEC) is a foundational analytical method to assess product purity of biological molecules. To ensure accurate and reproducible data that meet regulatory agency standards, it is critical to monitor the chromatographic column with efficient and continuous approaches. In this study, 19 SEC columns (Waters Acquity BEH200) were evaluated using an in-house monoclonal antibody made at Regeneron. System suitability parameters (SSPs) were used to monitor the performance of the SEC assay, including USP resolution, USP plate count, USP tailing factor, asymmetry factor, elution time, peak width, and peak height. A general linear model was built and revealed that elution time, peak width, asymmetry factor, and tailing factor increased with injection number, while peak height, resolution, and plate count decreased. After 1000 injections, tailing factor and peak width increased by more than 10%, while resolution and plate count decreased by more than 10% from their respective starting values.
Collapse
Affiliation(s)
- Kai-Wei Wu
- Department of Pharmaceutics and Drug Delivery, The University of Mississippi, University, MS 38677, USA
| | - Tse-Hong Chen
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA.
| | - Teng-Chieh Yang
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Shao-Chun Wang
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Mohammed Shameem
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| | - Kenneth S Graham
- Formulation Development Group, Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Rd, Tarrytown, NY 10591, USA
| |
Collapse
|
45
|
Kibria MG, Shiwaku Y, Brindha S, Kuroda Y. Biophysical and biochemical nature of amorphous protein oligomers determines the strength of immune response and the generation of T-cell memory. FEBS J 2023; 290:4712-4725. [PMID: 37287403 DOI: 10.1111/febs.16884] [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: 01/31/2023] [Revised: 04/26/2023] [Accepted: 06/06/2023] [Indexed: 06/09/2023]
Abstract
Here, we used domain 3 of dengue virus serotype 3 envelope protein (D3ED3), a natively folded globular low-immunogenicity protein, to ask whether the biophysical nature of amorphous oligomers can affect immunogenicity. We prepared nearly identical 30 ~ 50 nm-sized amorphous oligomers in five distinct ways and looked at any correlation between their biophysical properties and immunogenicity. One oligomer type was produced using our SCP tag (solubility controlling peptide) made of 5 isoleucines (C5I). The others were prepared by miss-shuffling the SS bonds (Ms), heating (Ht), stirring (St) and freeze-thaw (FT). Dynamic light scattering showed that all five formulations contained oligomers of approximately identical sizes with hydrodynamic radii (Rh) between 30 and 55 nm. Circular dichroism (cd) indicated that the secondary structure content of oligomers formed by stirring and freeze-thaw was essentially identical to that of the native monomeric D3ED3. The secondary structure content of the Ms showed moderate changes, whereas the C5I and heat-induced (Ht) oligomers exhibited a significant change. The Ms contained D3ED3 with intermolecular SS bonds as assessed by nonreducing size exclusion chromatography (SEC). Immunization in JcL:ICR mice showed that both C5I and Ms significantly increased the anti-D3ED3 IgG titre. Ht, St and FT were only mildly immunogenic, similar to the monomeric D3ED3. Cell surface CD marker analysis by flow cytometry confirmed that immunization with Ms generated a strong central and effector T-cell memory. Our observations indeed suggest that controlled oligomerization can provide a new, adjuvant-free method for increasing a protein's immunogenicity, yielding a potentially powerful platform for protein-based (subunit) vaccines.
Collapse
Affiliation(s)
- Md Golam Kibria
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Koganei-shi, Japan
| | - Yukari Shiwaku
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Koganei-shi, Japan
| | - Subbaian Brindha
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Koganei-shi, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Fuchu-Shi, Japan
| | - Yutaka Kuroda
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Koganei-shi, Japan
| |
Collapse
|
46
|
Pang KT, Yang YS, Zhang W, Ho YS, Sormanni P, Michaels TCT, Walsh I, Chia S. Understanding and controlling the molecular mechanisms of protein aggregation in mAb therapeutics. Biotechnol Adv 2023; 67:108192. [PMID: 37290583 DOI: 10.1016/j.biotechadv.2023.108192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/09/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
In antibody development and manufacturing, protein aggregation is a common challenge that can lead to serious efficacy and safety issues. To mitigate this problem, it is important to investigate its molecular origins. This review discusses (1) our current molecular understanding and theoretical models of antibody aggregation, (2) how various stress conditions related to antibody upstream and downstream bioprocesses can trigger aggregation, and (3) current mitigation strategies employed towards inhibiting aggregation. We discuss the relevance of the aggregation phenomenon in the context of novel antibody modalities and highlight how in silico approaches can be exploited to mitigate it.
Collapse
Affiliation(s)
- Kuin Tian Pang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore; School of Chemistry, Chemical Engineering, and Biotechnology, Nanyang Technology University, Singapore
| | - Yuan Sheng Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Wei Zhang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ying Swan Ho
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Pietro Sormanni
- Chemistry of Health, Yusuf Hamied Department of Chemistry, University of Cambridge, United Kingdom
| | - Thomas C T Michaels
- Department of Biology, Institute of Biochemistry, ETH Zurich, Otto-Stern-Weg 3, 8093 Zurich, Switzerland; Bringing Materials to Life Initiative, ETH Zurich, Switzerland
| | - Ian Walsh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Sean Chia
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore.
| |
Collapse
|
47
|
Porębska N, Ciura K, Chorążewska A, Zakrzewska M, Otlewski J, Opaliński Ł. Multivalent protein-drug conjugates - An emerging strategy for the upgraded precision and efficiency of drug delivery to cancer cells. Biotechnol Adv 2023; 67:108213. [PMID: 37453463 DOI: 10.1016/j.biotechadv.2023.108213] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/20/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
With almost 20 million new cases per year, cancer constitutes one of the most important challenges for public health systems. Unlike traditional chemotherapy, targeted anti-cancer strategies employ sophisticated therapeutics to precisely identify and attack cancer cells, limiting the impact of drugs on healthy cells and thereby minimizing the unwanted side effects of therapy. Protein drug conjugates (PDCs) are a rapidly growing group of targeted therapeutics, composed of a cancer-recognition factor covalently coupled to a cytotoxic drug. Several PDCs, mainly in the form of antibody-drug conjugates (ADCs) that employ monoclonal antibodies as cancer-recognition molecules, are used in the clinic and many PDCs are currently in clinical trials. Highly selective, strong and stable interaction of the PDC with the tumor marker, combined with efficient, rapid endocytosis of the receptor/PDC complex and its subsequent effective delivery to lysosomes, is critical for the efficacy of targeted cancer therapy with PDCs. However, the bivalent architecture of contemporary clinical PDCs is not optimal for tumor receptor recognition or PDCs internalization. In this review, we focus on multivalent PDCs, which represent a rapidly evolving and highly promising therapeutics that overcome most of the limitations of current bivalent PDCs, enhancing the precision and efficiency of drug delivery to cancer cells. We present an expanding set of protein scaffolds used to generate multivalent PDCs that, in addition to folding into well-defined multivalent molecular structures, enable site-specific conjugation of the cytotoxic drug to ensure PDC homogeneity. We provide an overview of the architectures of multivalent PDCs developed to date, emphasizing their efficacy in the targeted treatment of various cancers.
Collapse
Affiliation(s)
- Natalia Porębska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Krzysztof Ciura
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Aleksandra Chorążewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Małgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Łukasz Opaliński
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland.
| |
Collapse
|
48
|
Liu GY, Zhang Z, Yan Y, Wang S, Li N. Discovery and Characterization of an Acid-Labile Serine-Lysine Cross-Link in Antibody High-Molecular-Weight Species Using a Multipronged Mass Spectrometry Approach. Anal Chem 2023; 95:13813-13821. [PMID: 37674418 PMCID: PMC10515106 DOI: 10.1021/acs.analchem.3c01602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023]
Abstract
Characterizing the cross-links responsible for the covalent high-molecular-weight (HMW) species in therapeutic monoclonal antibodies (mAbs) is of great importance as it not only provides a framework for risk assessment but also offers insights for process improvement. However, owing to the complexity and low abundance, identification of novel and unknown cross-links in mAb products can be very challenging. Here, applying a multipronged MS-based approach, we report the discovery of a novel covalent cross-link formed via an imine bond between lysine and serine residues. In particular, this Ser-Lys cross-link was found to be acid-labile and can be easily overlooked by conventional LC-MS techniques operated at low pH. It is worth noting that although imine-based cross-link has been previously reported in collagen protein cross-linking, this is the first time that a Ser-Lys cross-link has been found in a mAb product that contributes to covalent HMW species formation.
Collapse
Affiliation(s)
- Gao-Yuan Liu
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Zhengqi Zhang
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Yuetian Yan
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Shunhai Wang
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| | - Ning Li
- Analytical Chemistry, Regeneron
Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591-6707, United States
| |
Collapse
|
49
|
Englert S, Mercier F, Pilling EA, Homer V, Habermehl C, Zimmermann S, Kan-Dobrosky N. Defining estimands for efficacy assessment in single arm phase 1b or phase 2 clinical trials in oncology early development. Pharm Stat 2023; 22:921-937. [PMID: 37403434 DOI: 10.1002/pst.2319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/06/2023]
Abstract
The addendum of the ICH E9 guideline on the statistical principles for clinical trials introduced the estimand framework. The framework is designed to strengthen the dialog between different stakeholders, to introduce greater clarity in the clinical trial objectives and to provide alignment between the estimand and statistical analysis. Estimand framework related publications thus far have mainly focused on randomized clinical trials. The intention of the Early Development Estimand Nexus (EDEN), a task force of the cross-industry Oncology Estimand Working Group (www.oncoestimand.org), is to apply it to single arms Phase 1b or Phase 2 trials designed to detect a treatment-related efficacy signal, typically measured by objective response rate. Key recommendations regarding the estimand attributes include that in a single arm early clinical trial, the treatment attribute should start when the first dose is received by the participant. Focusing on the estimation of an absolute effect, the population-level summary measure should reflect only the property used for the estimation. Another major component introduced in the ICH E9 addendum is the definition of intercurrent events and the associated possible ways to handle them. Different strategies reflect different clinical questions of interest that can be answered based on the journeys an individual subject can take during a trial. We provide detailed strategy recommendations for intercurrent events typically seen in early-stage oncology. We highlight where implicit assumptions should be made transparent as whenever follow-up is suspended, a while-on-treatment strategy is implied.
Collapse
Affiliation(s)
- Stefan Englert
- Statistical Modeling & Methodology, Janssen R&D, Janssen-Cilag GmbH, Neuss, Germany
| | - François Mercier
- Biostatistics, Roche Innovation Center Basel, F Hoffmann-La Roche AG, Basel, Switzerland
| | | | - Victoria Homer
- Cancer Research (UK) Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Christina Habermehl
- Global Biostatistics, The healthcare Business of Merck KgaA, Darmstadt, Germany
| | - Stefan Zimmermann
- Early Clinical Development Oncology, Roche Innovation Center Zurich, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Natalia Kan-Dobrosky
- Statistical Science, PPD, Part of Thermo Fisher Scientific, Wilmington, North Carolina, USA
| |
Collapse
|
50
|
Chen W, Klemm D, Gregoritza K, Satya Krishna Kishore R, Olaf Stracke J, Wurth C, Pinto C, Sancho Oltra N. Screening techniques for monitoring the sub-visible particle formation of free fatty acids in biopharmaceuticals. Eur J Pharm Biopharm 2023; 190:242-247. [PMID: 37524212 DOI: 10.1016/j.ejpb.2023.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
Free fatty acid (FFA) particles that originate from the enzymatic hydrolysis of polysorbate (PS) via co-purified host cell proteins generally appear abruptly in drug products during real-time (long-term) storage. Efforts were taken to understand the kinetics of FFA particle formation, aiming for a mitigation strategy. However, it is rather challenging particularly in the sub-visible particle (SVP) range, due to either the insufficient sensitivity of the analytical techniques used or the interference of the formulation matrices of proteinaceous drug products. In this study, we examined the feasibility of Raman microscopy, backgrounded membrane imaging (BMI) and total holographic characterization (THC) on the detection of FFA sub-visible particles (SVPs). The results indicate that THC is the most sensitive technique to track their occurrence during the course of PS hydrolysis. Moreover, with this technique we are able to distinguish different stages of FFA particle formation in the medium. In addition, a real time stability study of a biopharmaceutical was analyzed, demonstrating the viability of THC to monitor SVPs in a real sample and correlate it to the visible particles (VPs) occurrence.
Collapse
Affiliation(s)
- Wei Chen
- F. Hoffmann-La Roche AG, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Denis Klemm
- F. Hoffmann-La Roche AG, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | | | | | - Jan Olaf Stracke
- F. Hoffmann-La Roche AG, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Christine Wurth
- F. Hoffmann-La Roche AG, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Cosimo Pinto
- F. Hoffmann-La Roche AG, Grenzacherstrasse 124, 4070 Basel, Switzerland.
| | - Nuria Sancho Oltra
- F. Hoffmann-La Roche AG, Grenzacherstrasse 124, 4070 Basel, Switzerland.
| |
Collapse
|