1
|
Ito T, Suzuki T, Sakai Y, Nishioka K, Itoh Y, Sakamoto K, Ikemura N, Matoba S, Kanda Y, Takagi J, Okamoto T, Tahara K, Hoshino A. Engineered ACE2 decoy in dry powder form for inhalation: A novel therapy for SARS-CoV-2 variants. Mol Ther Methods Clin Dev 2025; 33:101459. [PMID: 40276779 PMCID: PMC12019485 DOI: 10.1016/j.omtm.2025.101459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
The persistent threat of SARS-CoV-2 and the emergence of new variants has prompted the development of a novel, easily administered modality that can overcome viral mutations. The engineered ACE2 decoy shows neutralizing activity comparable to monoclonal antibodies and is broadly effective against SARS-CoV-2 variants and ACE2-utilizing sarbecoviruses. In addition to intravenous administration, this decoy has shown antiviral efficacy through nebulized aerosol inhalation in murine and primate models, offering a dose-sparing advantage. Clinically, dry powder formulation is ideal for convenience and storage but poses challenges for protein biologics. This study developed a freeze-dried spray formulation of the ACE2 decoy for inhalation. The trehalose and leucine-based excipient maintained neutralizing activity and prevented aggregate formation. The dry powder showed aerodynamic distribution from bronchi to alveoli, aiding protection against SARS-CoV-2 infections. Neutralizing activity, structural stability, and powder dispersibility were preserved after 6 months of storage. In a mouse model of SARS-CoV-2 infection, significant reductions in viral replication and lung pathology were observed with intratracheal administration 24 h post-infection. The ACE2 decoy retained activity against recent JN.1 and current KP.3 strains, confirming its robust efficacy against viral mutations. This ACE2 decoy powder inhalant is a self-administered, next-generation treatment addressing the ongoing immune-evading evolution of SARS-CoV-2.
Collapse
Affiliation(s)
- Takaaki Ito
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tatsuya Suzuki
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Yusuke Sakai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| | - Keisuke Nishioka
- Department of Infectious Diseases, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yumi Itoh
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kentarou Sakamoto
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Nariko Ikemura
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Junichi Takagi
- Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| | - Toru Okamoto
- Department of Microbiology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
- Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kohei Tahara
- Laboratory of Pharmaceutical Engineering, Gifu Pharmaceutical University, Gifu 501-1196, Japan
- Laboratory of Nanofiber Technology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Atsushi Hoshino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
2
|
Kopp KT, Beer MD, Voorspoels J, Lysebetten DV, den Mooter GV. The value of spray drying as stabilization process for proteins. Int J Pharm 2025; 674:125422. [PMID: 40057212 DOI: 10.1016/j.ijpharm.2025.125422] [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: 01/08/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/17/2025]
Abstract
Protein stability in solution state is often poor due to the intrinsic instability of proteins. A solution is to solidify them by using techniques like freeze or spray drying (SD). To shield therapeutic proteins from stress (e.g., heat or shear stress) related to the solidification process, suitable buffers and excipients are added during formulation development. In this work, buffers and excipients were identified for the stabilization of three protein model compounds (BSA, IgG and lysozyme) in solution state using a design of experiments (DoE) approach based on screening results from differential scanning fluorimetry (DSF) combined with static light scattering (SLS). The aim was to investigate whether it is possible to predict protein stability in solid state using data from protein stabilization in solution state according to DSF/SLS. Therefore, three concepts per protein were analyzed after SD, two of which were expected to stabilize the protein, and one less stabilizing and compared these results to screening results obtained in solution state. Analytical techniques prior to and post SD were reversed-phase and size-exclusion chromatography (RPC and SEC, respectively), dynamic light scattering (DLS), UV and circular dichroism (CD). Furthermore, yield and residual moisture were analyzed. BSA and lysozyme showed high stability during SD and therefore only minor changes were observed. IgG was more affected by solidification which partly resulted in a loss of more than 15 % of the initial protein concentration in comparison to before SD. In future studies, the use of analytical techniques that do not require reconstitution would give additional value.
Collapse
Affiliation(s)
- Katharina Tatjana Kopp
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium; Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49, 3000 Leuven, Belgium
| | - Maarten De Beer
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium
| | - Jody Voorspoels
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium
| | | | - Guy Van den Mooter
- Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
3
|
Ioannou Sartzi M, Drettas D, Stramarkou M, Krokida M. A Comprehensive Review of the Latest Trends in Spray Freeze Drying and Comparative Insights with Conventional Technologies. Pharmaceutics 2024; 16:1533. [PMID: 39771512 PMCID: PMC11679755 DOI: 10.3390/pharmaceutics16121533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Spray freeze drying (SFD) represents an emerging drying technique designed to produce a wide range of pharmaceuticals, foods, and active components with high quality and enhanced stability due to their unique structural characteristics. This method combines the advantages of the well-established techniques of freeze drying (FD) and spray drying (SD) while overcoming their challenges related to high process temperatures and durations. This is why SFD has experienced steady growth in recent years regarding not only the research interest, which is reflected by the increasing number of literature articles, but most importantly, the expanded market adoption, particularly in the pharmaceutical sector. Despite its potential, the high initial investment costs and complex operational requirements may hinder its growth. This paper provides a comprehensive review of the SFD technology, highlighting its advantages over conventional drying techniques and presenting its latest applications focused on pharmaceuticals. It also offers a thorough examination of the principles and the various parameters influencing the process for a better understanding and optimization of the process according to the needs of the final product. Finally, the current limitations of SFD are discussed, and future directions for addressing the economic and technical barriers are provided so that SFD can be widely industrialized, unlocking its full potential for diverse applications.
Collapse
Affiliation(s)
| | | | - Marina Stramarkou
- Laboratory of Process Analysis and Design, School of Chemical Engineering, National Technical University of Athens, 9 Iroon Polytechneiou St. Zografou Campus, 15780 Athens, Greece; (M.I.S.); (D.D.); (M.K.)
| | | |
Collapse
|
4
|
Kopp KT, Saerens L, Voorspoels J, Van den Mooter G. Solidification and oral delivery of biologics to the colon- A review. Eur J Pharm Sci 2023; 190:106523. [PMID: 37429482 DOI: 10.1016/j.ejps.2023.106523] [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: 02/01/2023] [Revised: 06/16/2023] [Accepted: 07/08/2023] [Indexed: 07/12/2023]
Abstract
The oral delivery of biologics such as therapeutic proteins, peptides and oligonucleotides for the treatment of colon related diseases has been the focus of increasing attention over the last years. However, the major disadvantage of these macromolecules is their degradation propensity in liquid state which can lead to the undesirable and complete loss of function. Therefore, to increase the stability of the biologic and reduce their degradation propensity, formulation techniques such as solidification can be performed to obtain a stable solid dosage form for oral administration. Due to their fragility, stress exerted on the biologic during solidification has to be reduced with the incorporation of stabilizing excipients into the formulation. This review focuses on the state-of-the-art solidification techniques required to obtain a solid dosage form for the oral delivery of biologics to the colon and the use of suitable excipients for adequate stabilization upon solidification. The solidifying processes discussed within this review are spray drying, freeze drying, bead coating and also other techniques such as spray freeze drying, electro spraying, vacuum- and supercritical fluid drying. Further, the colon as site of absorption in both healthy and diseased state is critically reviewed and possible oral delivery systems for biologics are discussed.
Collapse
Affiliation(s)
- Katharina Tatjana Kopp
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium; Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49, 3000 Leuven, Belgium
| | - Lien Saerens
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium
| | - Jody Voorspoels
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium
| | - Guy Van den Mooter
- Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
5
|
Recent progress in drying technologies for improving the stability and delivery efficiency of biopharmaceuticals. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2023; 53:35-57. [PMID: 36568503 PMCID: PMC9768793 DOI: 10.1007/s40005-022-00610-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Background Most biopharmaceuticals are developed in liquid dosage forms that are less stable than solid forms. To ensure the stability of biopharmaceuticals, it is critical to use an effective drying technique in the presence of an appropriate stabilizing excipient. Various drying techniques are available for this purpose, such as freeze drying or lyophilization, spray drying, spray freeze-drying, supercritical fluid drying, particle replication in nonwetting templates, and fluidized bed drying. Area covered In this review, we discuss drying technologies and their applications in the production of stable solid-state biopharmaceuticals, providing examples of commercially available products or clinical trial formulations. Alongside this, we also review how different analytical methods may be utilized in the evaluation of aerosol performance and powder characteristics of dried protein powders. Finally, we assess the protein integrity in terms of conformational and physicochemical stability and biological activity. Expert opinion With the aim of treating either infectious respiratory diseases or systemic disorders, inhaled biopharmaceuticals reduce both therapeutic dose and cost of therapy. Drying methods in the presence of optimized protein/stabilizer combinations, produce solid dosage forms of proteins with greater stability. A suitable drying method was chosen, and the process parameters were optimized based on the route of protein administration. With the ongoing trend of addressing deficiencies in biopharmaceutical production, developing new methods to replace conventional drying methods, and investigating novel excipients for more efficient stabilizing effects, these products have the potential to dominate the pharmaceutical industry in the future.
Collapse
|
6
|
Son JW, Son JM, Hur KH, Lee W, Song I, Na DH. Application of isothermal chemical denaturation to early‐stage formulation development of fibrinogen. B KOREAN CHEM SOC 2022. [DOI: 10.1002/bkcs.12664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Jae Woon Son
- GC Biopharma Yongin Republic of Korea
- College of Pharmacy Kyungpook National University Daegu Republic of Korea
| | - Jong Mun Son
- GC Biopharma Yongin Republic of Korea
- College of Pharmacy Chung‐Ang University Seoul Republic of Korea
| | - Ki Ho Hur
- GC Biopharma Yongin Republic of Korea
- College of Pharmacy Chungbuk National University Cheongju South Korea
| | - Wonhwa Lee
- Department of Chemistry Sungkyunkwan University Suwon Republic of Korea
| | - Im‐Sook Song
- College of Pharmacy Kyungpook National University Daegu Republic of Korea
| | - Dong Hee Na
- College of Pharmacy Chung‐Ang University Seoul Republic of Korea
| |
Collapse
|
7
|
Spray Freeze Drying of Biologics: A Review and Applications for Inhalation Delivery. Pharm Res 2022; 40:1115-1140. [DOI: 10.1007/s11095-022-03442-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022]
|
8
|
Ricardo PC, Serudo RL, Ţălu Ş, Lamarão CV, da Fonseca Filho HD, de Araújo Bezerra J, Sanches EA, Campelo PH. Encapsulation of Bromelain in Combined Sodium Alginate and Amino Acid Carriers: Experimental Design of Simplex-Centroid Mixtures for Digestibility Evaluation. Molecules 2022; 27:6364. [PMID: 36234901 PMCID: PMC9570880 DOI: 10.3390/molecules27196364] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Bromelain has potential as an analgesic, an anti-inflammatory, and in cancer treatments. Despite its therapeutic effects, this protein undergoes denaturation when administered orally. Microencapsulation processes have shown potential in protein protection and as controlled release systems. Thus, this paper aimed to develop encapsulating systems using sodium alginate as a carrier material and positively charged amino acids as stabilizing agents for the controlled release of bromelain in in vitro tests. The systems were produced from the experimental design of centroid simplex mixtures. Characterizations were performed by FTIR showing that bromelain was encapsulated in all systems. XRD analyses showed that the systems are semi-crystalline solids and through SEM analysis the morphology of the formed systems followed a pattern of rough microparticles. The application of statistical analysis showed that the systems presented behavior that can be evaluated by quadratic and special cubic models, with a p-value < 0.05. The interaction between amino acids and bromelain/alginate was evaluated, and free bromelain showed a reduction of 74.0% in protein content and 23.6% in enzymatic activity at the end of gastric digestion. Furthermore, a reduction of 91.6% of protein content and 65.9% of enzymatic activity was observed at the end of intestinal digestion. The Lis system showed better interaction due to the increased stability of bromelain in terms of the amount of proteins (above 63% until the end of the intestinal phase) and the enzymatic activity of 89.3%. Thus, this study proposes the development of pH-controlled release systems aiming at increasing the stability and bioavailability of bromelain in intestinal systems.
Collapse
Affiliation(s)
- Philipi Cavalcante Ricardo
- Graduate Program in Materials Science and Engineering (PPGCEM), Federal University of Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - Ricardo Lima Serudo
- Higher School of Technology (EST), State University of Amazonas (UEA), Av. Djalma Batista 2470, Manaus 69050-300, AM, Brazil
| | - Ştefan Ţălu
- The Directorate of Research, Development and Innovation Management (DMCDI), Technical University of Cluj-Napoca, 15 Constantin Daicoviciu St., 400020 Cluj-Napoca, Romania
| | - Carlos Victor Lamarão
- School of Agrarian Science, Federal University of Amazonas, Manaus 69067-005, AM, Brazil
| | - Henrique Duarte da Fonseca Filho
- Graduate Program in Materials Science and Engineering (PPGCEM), Federal University of Amazonas (UFAM), Manaus 69067-005, AM, Brazil
- Laboratory of Synthesis of Nanomaterials and Nanoscopy (LSNN), Department of Physics, Federal University of Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - Jaqueline de Araújo Bezerra
- Federal Institute of Education, Science and Technology of Amazonas (IFAM), IFAM Analytical Center, Manaus Centro Campus, Manaus 69067-005, AM, Brazil
| | - Edgar Aparecido Sanches
- Graduate Program in Materials Science and Engineering (PPGCEM), Federal University of Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - Pedro Henrique Campelo
- Graduate Program in Materials Science and Engineering (PPGCEM), Federal University of Amazonas (UFAM), Manaus 69067-005, AM, Brazil
- Department of Food Technology, Federal University of Viçosa (UFV), Viçosa 36570-900, MG, Brazil
| |
Collapse
|
9
|
Kim SH, Yoo HJ, Park EJ, Lee W, Na DH. Impact of buffer concentration on the thermal stability of immunoglobulin G. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00587-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
10
|
Nano Differential Scanning Fluorimetry-Based Thermal Stability Screening and Optimal Buffer Selection for Immunoglobulin G. Pharmaceuticals (Basel) 2021; 15:ph15010029. [PMID: 35056086 PMCID: PMC8778976 DOI: 10.3390/ph15010029] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/12/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Nano differential scanning fluorimetry (nanoDSF) is a high-throughput protein stability screening technique that simultaneously monitors protein unfolding and aggregation properties. The thermal stability of immunoglobulin G (IgG) was investigated in three different buffers (sodium acetate, sodium citrate, and sodium phosphate) ranging from pH 4 to 8. In all three buffers, the midpoint temperature of thermal unfolding (Tm) showed a tendency to increase as the pH increased, but the aggregation propensity was different depending on the buffer species. The best stability against aggregation was obtained in the sodium acetate buffers below pH 4.6. On the other hand, IgG in the sodium citrate buffer had higher aggregation and viscosity than in the sodium acetate buffer at the same pH. Difference of aggregation between acetate and citrate buffers at the same pH could be explained by a protein-protein interaction study, performed with dynamic light scattering, which suggested that intermolecular interaction is attractive in citrate buffer but repulsive in acetate buffer. In conclusion, this study indicates that the sodium acetate buffer at pH 4.6 is suitable for IgG formulation, and the nanoDSF method is a powerful tool for thermal stability screening and optimal buffer selection in antibody formulations.
Collapse
|
11
|
Cun D, Zhang C, Bera H, Yang M. Particle engineering principles and technologies for pharmaceutical biologics. Adv Drug Deliv Rev 2021; 174:140-167. [PMID: 33845039 DOI: 10.1016/j.addr.2021.04.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/21/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
The global market of pharmaceutical biologics has expanded significantly during the last few decades. Currently, pharmaceutical biologic products constitute an indispensable part of the modern medicines. Most pharmaceutical biologic products are injections either in the forms of solutions or lyophilized powders because of their low oral bioavailability. There are certain pharmaceutical biologic entities formulated into particulate delivery systems for the administration via non-invasive routes or to achieve prolonged pharmaceutical actions to reduce the frequency of injections. It has been well documented that the design of nano- and microparticles via various particle engineering technologies could render pharmaceutical biologics with certain benefits including improved stability, enhanced intracellular uptake, prolonged pharmacological effect, enhanced bioavailability, reduced side effects, and improved patient compliance. Herein, we review the principles of the particle engineering technologies based on bottom-up approach and present the important formulation and process parameters that influence the critical quality attributes with some mathematical models. Subsequently, various nano- and microparticle engineering technologies used to formulate or process pharmaceutical biologic entities are reviewed. Lastly, an array of commercialized products of pharmaceutical biologics accomplished based on various particle engineering technologies are presented and the challenges in the development of particulate delivery systems for pharmaceutical biologics are discussed.
Collapse
Affiliation(s)
- Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016 Shenyang, China
| | - Chengqian Zhang
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Hriday Bera
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016 Shenyang, China
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016 Shenyang, China; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
12
|
Chen Y, Mutukuri TT, Wilson NE, Zhou QT. Pharmaceutical protein solids: Drying technology, solid-state characterization and stability. Adv Drug Deliv Rev 2021; 172:211-233. [PMID: 33705880 PMCID: PMC8107147 DOI: 10.1016/j.addr.2021.02.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/18/2021] [Accepted: 02/22/2021] [Indexed: 01/30/2023]
Abstract
Despite the boom in biologics over the past decade, the intrinsic instability of these large molecules poses significant challenges to formulation development. Almost half of all pharmaceutical protein products are formulated in the solid form to preserve protein native structure and extend product shelf-life. In this review, both traditional and emerging drying techniques for producing protein solids will be discussed. During the drying process, various stresses can impact the stability of protein solids. However, understanding the impact of stress on protein product quality can be challenging due to the lack of reliable characterization techniques for biological solids. Both conventional and advanced characterization techniques are discussed including differential scanning calorimetry (DSC), solid-state Fourier transform infrared spectrometry (ssFTIR), solid-state fluorescence spectrometry, solid-state hydrogen deuterium exchange (ssHDX), solid-state nuclear magnetic resonance (ssNMR) and solid-state photolytic labeling (ssPL). Advanced characterization tools may offer mechanistic investigations into local structural changes and interactions at higher resolutions. The continuous exploration of new drying techniques, as well as a better understanding of the effects caused by different drying techniques in solid state, would advance the formulation development of biological products with superior quality.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Tarun Tejasvi Mutukuri
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Nathan E Wilson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
13
|
Novel formulations and drug delivery systems to administer biological solids. Adv Drug Deliv Rev 2021; 172:183-210. [PMID: 33705873 DOI: 10.1016/j.addr.2021.02.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Recent advances in formulation sciences have expanded the previously limited design space for biological modalities, including peptide, protein, and vaccine products. At the same time, the discovery and application of new modalities, such as cellular therapies and gene therapies, have presented formidable challenges to formulation scientists. We explore these challenges and highlight the opportunities to overcome them through the development of novel formulations and drug delivery systems as biological solids. We review the current progress in both industry and academic laboratories, and we provide expert perspectives in those settings. Formulation scientists have made a tremendous effort to accommodate the needs of these novel delivery routes. These include stability-preserving formulations and dehydration processes as well as dosing regimes and dosage forms that improve patient compliance.
Collapse
|
14
|
Keyhan shokouh M, Faghihi H, Darabi M, Mirmoeini M, Vatanara A. Formulation and evaluation of inhalable microparticles of Rizatriptan Benzoate processed by spray freeze-drying. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
15
|
Nishimura S, Murakami Y. Precise Control of the Surface and Internal Morphologies of Porous Particles Prepared Using a Spontaneous Emulsification Method. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:3075-3085. [PMID: 33657324 DOI: 10.1021/acs.langmuir.0c03311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Porous particles with controllable surface and internal morphologies were successfully prepared by a "one-step mechanical emulsification" technique via the control of spontaneous emulsification where self-emulsification is followed by mechanical emulsification. The morphological changes in the porous particles were determined not by the preparation conditions of the water-in-oil-in-water (w/o/w) emulsion but by the proportion of solvents that favors the stabilization of the spontaneously prepared water-in-oil (w/o) emulsion droplets acting as porogens. The proposed method for controlling the morphology of the porous particles could be applied to all particle-preparation systems based on emulsion-solvent evaporation using organic solvents. The methodology for the morphological control of porous particles independent of the concentration or composition of the polymer is considered valuable for future investigations into the aerodynamic performance and drug-release behavior of biomedical porous particles with complex shapes.
Collapse
Affiliation(s)
- Shinnosuke Nishimura
- Department of Organic and Polymer Materials Chemistry, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Yoshihiko Murakami
- Department of Organic and Polymer Materials Chemistry, Faculty of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| |
Collapse
|
16
|
Formulating monoclonal antibodies as powders for reconstitution at high concentration using spray-drying: Trehalose/amino acid combinations as reconstitution time reducing and stability improving formulations. Eur J Pharm Biopharm 2020; 156:131-142. [DOI: 10.1016/j.ejpb.2020.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/11/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
|
17
|
Spray Freeze-Drying as a Solution to Continuous Manufacturing of Pharmaceutical Products in Bulk. Processes (Basel) 2020. [DOI: 10.3390/pr8060709] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pharmaceutical manufacturing is evolving from traditional batch processes to continuous ones. The new global competition focused on throughput and quality of drug products is certainly the driving force behind this transition which, thus, represents the new challenge of pharmaceutical manufacturing and hence of lyophilization as a downstream operation. In this direction, the present review deals with the most recent technologies, based on spray freeze-drying, that can achieve this objective. It provides a comprehensive overview of the physics behind this process and of the most recent equipment design.
Collapse
|
18
|
Emami F, Vatanara A, Vakhshiteh F, Kim Y, Kim TW, Na DH. Amino acid-based stable adalimumab formulation in spray freeze-dried microparticles for pulmonary delivery. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101249] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
19
|
Wang W, Ohtake S. Science and art of protein formulation development. Int J Pharm 2019; 568:118505. [PMID: 31306712 DOI: 10.1016/j.ijpharm.2019.118505] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023]
Abstract
Protein pharmaceuticals have become a significant class of marketed drug products and are expected to grow steadily over the next decade. Development of a commercial protein product is, however, a rather complex process. A critical step in this process is formulation development, enabling the final product configuration. A number of challenges still exist in the formulation development process. This review is intended to discuss these challenges, to illustrate the basic formulation development processes, and to compare the options and strategies in practical formulation development.
Collapse
Affiliation(s)
- Wei Wang
- Biological Development, Bayer USA, LLC, 800 Dwight Way, Berkeley, CA 94710, United States.
| | - Satoshi Ohtake
- Pharmaceutical Research and Development, Pfizer Biotherapeutics Pharmaceutical Sciences, Chesterfield, MO 63017, United States
| |
Collapse
|
20
|
Poly(lactic acid)/poly(lactic-co-glycolic acid) particulate carriers for pulmonary drug delivery. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-019-00443-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
21
|
Mirfakhraei Y, Faghihi H, Zade AHM, Darabi M, Vatanara A. Optimization of Stable IgG Formulation Containing Amino Acids and Trehalose During Freeze-Drying and After Storage: a Central Composite Design. AAPS PharmSciTech 2019; 20:154. [PMID: 30919164 DOI: 10.1208/s12249-019-1322-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 01/26/2019] [Indexed: 01/03/2023] Open
Abstract
The physical and structural stability of freeze-dried immunoglobulin G (IgG) were examined by applying trehalose and amino acids (glycine, phenylalanine, and serine). The efficacy of amino acids was statistically compared considering their side-chain characteristics. The amount of amino acids (X1) and trehalose (X2) was considered as independent variables. Size exclusion chromatography (SEC-HPLC) was utilized to calculate the soluble aggregates, as dependent variables. The amounts of excipients were optimized through the central composite design (CCD). The beta-sheet conformation of IgG was quantified by Fourier transform infrared spectroscopy (FTIR). Thermal behavior and molecular integrity of IgG were evaluated by differential scanning calorimetry (DSC) and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Optimized formulations of powders were as follows: 24.5 mg serine-139.5 mg trehalose, 14 mg glycine-118 mg Trehalose, and 25 mg phenylalanine-139.5 mg trehalose. The amounts of soluble aggregates after processing were 0, 4.50, and 2.20%, respectively. The corresponding induced aggregates following storage conditions were 1.02, 7.0, and 3.70%. In all preparations, there were no detectable fragments. The native conformation of IgG was well preserved in the presence of amino acids. Excluding the glycine-based sample with minor endotherm at about 45°C, serine and phenylalanine incorporating powders were fully amorphous at examination temperatures. Trehalose was more potent than the amino acids in the stabilization of IgG. Serine was the most effective amino acid; phenylalanine and glycine were the next ones, respectively. Glycine crystallization was assumed to have accounted for low stabilization capability. The statistically synergistic phenomenon was only observed in the co-application of trehalose and phenylalanine. Graphical abstract.
Collapse
|
22
|
Dutta S, Moses JA, Anandharamakrishnan C. Modern frontiers and applications of spray‐freeze‐drying in design of food and biological supplements. J FOOD PROCESS ENG 2018. [DOI: 10.1111/jfpe.12881] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Sayantani Dutta
- Computational Modeling and Nano Scale Processing UnitIndian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India Thanjavur Tamil Nadu India
| | - J. A. Moses
- Computational Modeling and Nano Scale Processing UnitIndian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India Thanjavur Tamil Nadu India
| | - C. Anandharamakrishnan
- Computational Modeling and Nano Scale Processing UnitIndian Institute of Food Processing Technology (IIFPT), Ministry of Food Processing Industries, Government of India Thanjavur Tamil Nadu India
| |
Collapse
|
23
|
Identification of D-Amino Acids in Light Exposed mAb Formulations. Pharm Res 2018; 35:238. [PMID: 30334107 DOI: 10.1007/s11095-018-2520-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/05/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE We previously demonstrated that D-amino acids can form as a result of photo-irradiation of a monoclonal antibody (mAb) at both λ = 254 nm and λ > 295 nm (λmax = 305 nm), likely via reversible hydrogen transfer reactions of intermediary thiyl radicals. Here, we investigate the role of various excipients (sucrose, glucose, L-Arg, L-Met and L-Leu) on D-amino acid formation, and specifically the distribution of D-amino acids in mAb monomers and aggregates present after light exposure. METHODS The mAb-containing formulations were photo-irradiated at λ = 254 nm and λmax = 305 nm, followed by fractionation of aggregate and monomer fractions using size exclusion chromatography. These aggregate and monomer fractions were subjected to hydrolysis and subsequent amino acid analysis. RESULTS Both aggregate and monomer fractions collected from all formulations showed the formation of D-Glu and D-Val, whereas the formation of D-Ala was limited to the aggregate fraction collected from an L-Arg-containing formulation. Interestingly, quantitative analysis revealed higher yields of D-amino acids in the L-Arg-containing formulation. CONCLUSIONS Generally, D-amino acids accumulated to similar extents in monomers and aggregates.
Collapse
|
24
|
Emami F, Vatanara A, Park EJ, Na DH. Drying Technologies for the Stability and Bioavailability of Biopharmaceuticals. Pharmaceutics 2018; 10:E131. [PMID: 30126135 PMCID: PMC6161129 DOI: 10.3390/pharmaceutics10030131] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 01/29/2023] Open
Abstract
Solid dosage forms of biopharmaceuticals such as therapeutic proteins could provide enhanced bioavailability, improved storage stability, as well as expanded alternatives to parenteral administration. Although numerous drying methods have been used for preparing dried protein powders, choosing a suitable drying technique remains a challenge. In this review, the most frequent drying methods, such as freeze drying, spray drying, spray freeze drying, and supercritical fluid drying, for improving the stability and bioavailability of therapeutic proteins, are discussed. These technologies can prepare protein formulations for different applications as they produce particles with different sizes and morphologies. Proper drying methods are chosen, and the critical process parameters are optimized based on the proposed route of drug administration and the required pharmacokinetics. In an optimized drying procedure, the screening of formulations according to their protein properties is performed to prepare a stable protein formulation for various delivery systems, including pulmonary, nasal, and sustained-release applications.
Collapse
Affiliation(s)
- Fakhrossadat Emami
- College of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran.
| | - Alireza Vatanara
- College of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran.
| | - Eun Ji Park
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea.
| | - Dong Hee Na
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea.
| |
Collapse
|