201
|
Gonzalez-Obeso C, Jane Hartzell E, Albert Scheel R, Kaplan DL. Delivering on the promise of recombinant silk-inspired proteins for drug delivery. Adv Drug Deliv Rev 2023; 192:114622. [PMID: 36414094 PMCID: PMC9812964 DOI: 10.1016/j.addr.2022.114622] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/06/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
Abstract
Effective drug delivery is essential for the success of a medical treatment. Polymeric drug delivery systems (DDSs) are preferred over systemic administration of drugs due to their protection capacity, directed release, and reduced side effects. Among the numerous polymer sources, silks and recombinant silks have drawn significant attention over the past decade as DDSs. Native silk is produced from a variety of organisms, which are then used as sources or guides of genetic material for heterologous expression or engineered designs. Recombinant silks bear the outstanding properties of natural silk, such as processability in aqueous solution, self-assembly, drug loading capacity, drug stabilization/protection, and degradability, while incorporating specific properties beneficial for their success as DDS, such as monodispersity and tailored physicochemical properties. Moreover, the on-demand inclusion of sequences that customize the DDS for the specific application enhances efficiency. Often, inclusion of a drug into a DDS is achieved by simple mixing or diffusion and stabilized by non-specific molecular interactions; however, these interactions can be improved by the incorporation of drug-binding peptide sequences. In this review we provide an overview of native sources for silks and silk sequences, as well as the design and formulation of recombinant silk biomaterials as drug delivery systems in a variety of formats, such as films, hydrogels, porous sponges, or particles.
Collapse
Affiliation(s)
- Constancio Gonzalez-Obeso
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, USA
| | - Emily Jane Hartzell
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, USA
| | - Ryan Albert Scheel
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, USA.
| |
Collapse
|
202
|
Anti-cancer Nanotechnology. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
203
|
Wang W, Jiang Y, Huang Z, Nguyen HVT, Liu B, Hartweg M, Shirakura M, Qin KP, Johnson JA. Discrete, Chiral Polymer-Insulin Conjugates. J Am Chem Soc 2022; 144:23332-23339. [PMID: 36126328 PMCID: PMC10440729 DOI: 10.1021/jacs.2c07382] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Polymer conjugation has been widely used to improve the stability and pharmacokinetics of therapeutic biomacromolecules; however, conventional methods to generate such conjugates often use disperse and/or achiral polymers with limited functionality. The heterogeneity of such conjugates may lead to manufacturing variability, poorly controlled biological performance, and limited ability to optimize structure-property relationships. Here, using insulin as a model therapeutic polypeptide, we introduce a strategy for the synthesis of polymer-protein conjugates based on discrete, chiral polymers synthesized through iterative exponential growth (IEG). These conjugates eliminate manufacturing variables originating from polymer dispersity and poorly controlled absolute configuration. Moreover, they offer tunable molecular features, such as conformational rigidity, that can be modulated to impact protein function, enabling faster or longer-lasting blood glucose responses in diabetic mice when compared to PEGylated insulin and the commercial insulin variant Lantus. Furthermore, IEG-insulin conjugates showed no signs of decreased activity, immunogenicity, or toxicity following repeat dosing. This work represents a significant step toward the synthesis of precise synthetic polymer-biopolymer conjugates and reveals that fine tuning of synthetic polymer structure may be used to optimize such conjugates in the future.
Collapse
Affiliation(s)
- Wencong Wang
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Yivan Jiang
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Zhihao Huang
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Hung V.-T. Nguyen
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Bin Liu
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Manuel Hartweg
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Masamichi Shirakura
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - K. Peter Qin
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Jeremiah A. Johnson
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
204
|
Han S, Wu J. Development of a Lysine-Based Poly(ester amide) Library with High Biosafety and a Finely Tunable Structure for Spatiotemporal-Controlled Protein Delivery. ACS APPLIED MATERIALS & INTERFACES 2022; 14:55944-55956. [PMID: 36503257 DOI: 10.1021/acsami.2c16492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
With the fast growth of protein therapeutics, efficient, precise, and universal delivery platforms are highly required. However, very few reports have discussed the progress of precisely spatiotemporal-controlled protein delivery. Therefore, a mini library of well-designed amino acid-based poly(ester amide)s derived from lysine (Lys-aaPEAs) has been developed. Lys-aaPEAs can interact with and encapsulate proteins into nanocomplexes via electrostatic interactions. The chemical structure of Lys-aaPEAs can be finely tuned by changing the type and molar ratio of the monomers. Studies of structure-function relationships reveal that the carbon chain length of diacid/diol segments, hydrophilicity, and electrical properties affect the polymer-protein interaction, cell-material interaction, and, therefore, the outcome of protein delivery. By modulating the structures of Lys-aaPEAs, the delivery systems could present customized physiochemical and biological properties and perform time- and space-specific protein release and delivery without causing any systematic toxicity. The screened systems exhibited prolonged hypoglycemic activity and superior biosafety in vivo, using insulin as a model protein and a mouse model bearing type 1 diabetes mellitus (T1DM). This work establishes a novel lysine-based polymer platform for spatiotemporal-controlled protein delivery and offers a paradigm of precise structure-function controllability for designing the next generation of polymers.
Collapse
Affiliation(s)
- Shuyan Han
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, P. R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, P. R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, P. R. China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P. R. China
| |
Collapse
|
205
|
Kapelner RA, Fisher RS, Elbaum-Garfinkle S, Obermeyer AC. Protein charge parameters that influence stability and cellular internalization of polyelectrolyte complex micelles. Chem Sci 2022; 13:14346-14356. [PMID: 36545145 PMCID: PMC9749388 DOI: 10.1039/d2sc00192f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Proteins are an important class of biologics, but there are several recurring challenges to address when designing protein-based therapeutics. These challenges include: the propensity of proteins to aggregate during formulation, relatively low loading in traditional hydrophobic delivery vehicles, and inefficient cellular uptake. This last criterion is particularly challenging for anionic proteins as they cannot cross the anionic plasma membrane. Here we investigated the complex coacervation of anionic proteins with a block copolymer of opposite charge to form polyelectrolyte complex (PEC) micelles for use as a protein delivery vehicle. Using genetically modified variants of the model protein green fluorescent protein (GFP), we evaluated the role of protein charge and charge localization in the formation and stability of PEC micelles. A neutral-cationic block copolymer, poly(oligoethylene glycol methacrylate-block-quaternized 4-vinylpyridine), POEGMA79-b-qP4VP175, was prepared via RAFT polymerization for complexation and microphase separation with the panel of engineered anionic GFPs. We found that isotropically supercharged proteins formed micelles at higher ionic strength relative to protein variants with charge localized to a polypeptide tag. We then studied GFP delivery by PEC micelles and found that they effectively delivered the protein cargo to mammalian cells. However, cellular delivery varied as a function of protein charge and charge distribution and we found an inverse relationship between the PEC micelle critical salt concentration and delivery efficiency. This model system has highlighted the potential of polyelectrolyte complexes to deliver anionic proteins intracellularly. Using this model system, we have identified requirements for the formation of PEC micelles that are stable at physiological ionic strength and that smaller protein-polyelectrolyte complexes effectively deliver proteins to Jurkat cells.
Collapse
Affiliation(s)
- Rachel A Kapelner
- Department of Chemical Engineering, Columbia University New York NY 10027 USA +1-212-853-1215
| | - Rachel S Fisher
- Department of Chemical Engineering, Columbia University New York NY 10027 USA +1-212-853-1215
- Structural Biology Initiative, CUNY Advanced Science Research Center New York NY USA
| | - Shana Elbaum-Garfinkle
- Structural Biology Initiative, CUNY Advanced Science Research Center New York NY USA
- PhD Programs in Biochemistry and Biology at the Graduate Center, City University of New York NY USA
| | - Allie C Obermeyer
- Department of Chemical Engineering, Columbia University New York NY 10027 USA +1-212-853-1215
| |
Collapse
|
206
|
Barrientos RC, Losacco GL, Azizi M, Wang H, Nguyen AN, Shchurik V, Singh A, Richardson D, Mangion I, Guillarme D, Regalado EL, Haidar Ahmad IA. Automated Hydrophobic Interaction Chromatography Screening Combined with In Silico Optimization as a Framework for Nondenaturing Analysis and Purification of Biopharmaceuticals. Anal Chem 2022; 94:17131-17141. [PMID: 36441925 DOI: 10.1021/acs.analchem.2c03453] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The mounting complexity of new modalities in the biopharmaceutical industry entails a commensurate level of analytical innovations to enable the rapid discovery and development of novel therapeutics and vaccines. Hydrophobic interaction chromatography (HIC) has become one of the widely preferred separation techniques for the analysis and purification of biopharmaceuticals under nondenaturing conditions. Inarguably, HIC method development remains very challenging and labor-intensive owing to the numerous factors that are typically optimized by a "hit-or-miss" strategy (e.g., the nature of the salt, stationary phase chemistry, temperature, mobile phase additive, and ionic strength). Herein, we introduce a new HIC method development framework composed of a fully automated multicolumn and multieluent platform coupled with in silico multifactorial simulation and integrated fraction collection for streamlined method screening, optimization, and analytical-scale purification of biopharmaceutical targets. The power and versatility of this workflow are showcased by a wide range of applications including trivial proteins, monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), oxidation variants, and denatured proteins. We also illustrate convenient and rapid HIC method development outcomes from the effective combination of this screening setup with computer-assisted simulations. HIC retention models were built using readily available LC simulator software outlining less than a 5% difference between experimental and simulated retention times with a correlation coefficient of >0.99 for pharmaceutically relevant multicomponent mixtures. In addition, we demonstrate how this approach paves the path for a straightforward identification of first-dimension HIC conditions that are combined with mass spectrometry (MS)-friendly reversed-phase liquid chromatography (RPLC) detection in the second dimension (heart-cutting two-dimensional (2D)-HIC-RPLC-diode array detector (DAD)-MS), enabling the analysis and purification of biopharmaceutical targets.
Collapse
Affiliation(s)
- Rodell C Barrientos
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Gioacchino Luca Losacco
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Mohammadmehdi Azizi
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Heather Wang
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Anh Nguyet Nguyen
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Vladimir Shchurik
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Andrew Singh
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Douglas Richardson
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Ian Mangion
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Davy Guillarme
- School of Pharmaceutical Sciences, University of Geneva, CMU, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 11 Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - Erik L Regalado
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Imad A Haidar Ahmad
- Analytical Research and Development, MRL, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| |
Collapse
|
207
|
Impact of N-Linked Glycosylation on Therapeutic Proteins. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248859. [PMID: 36557993 PMCID: PMC9781892 DOI: 10.3390/molecules27248859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022]
Abstract
Therapeutic proteins have unique advantages over small-molecule drugs in the treatment of various diseases, such as higher target specificity, stronger pharmacological efficacy and relatively low side effects. These advantages make them increasingly valued in drug development and clinical practice. However, although highly valued, the intrinsic limitations in their physical, chemical and pharmacological properties often restrict their wider applications. As one of the most important post-translational modifications, glycosylation has been shown to exert positive effects on many properties of proteins, including molecular stability, and pharmacodynamic and pharmacokinetic characteristics. Glycoengineering, which involves changing the glycosylation patterns of proteins, is therefore expected to be an effective means of overcoming the problems of therapeutic proteins. In this review, we summarize recent efforts and advances in the glycoengineering of erythropoietin and IgG monoclonal antibodies, with the goals of illustrating the importance of this strategy in improving the performance of therapeutic proteins and providing a brief overview of how glycoengineering is applied to protein-based drugs.
Collapse
|
208
|
Melodia D, Di Pietro Z, Cao C, Stenzel MH, Chapman R. Traceless pH-Sensitive Antibody Conjugation Inspired by Citraconic Anhydride. Biomacromolecules 2022; 23:5322-5329. [PMID: 36395470 DOI: 10.1021/acs.biomac.2c01125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We introduce a pH-sensitive amide bond, inspired by citraconic anhydride, for the reversible conjugation of polymers to the lysine residues of proteins and antibodies. The pH sensitivity arises from a conformation lock at the end of the polymer, which we introduce by means of a Diels-Alder reaction, that positions a carboxylic acid close to the amide after conjugation occurs. The amide is stable over weeks at pH 7.4 but sensitive to hydrolysis at pH 5.5 and below, returning the amine to its original state. The pH sensitivity can be tuned by positioning secondary amide groups nearby. We use this approach to PEGylate an antibody to human serum albumin at high dilution and demonstrate successful recovery of the activity after hydrolysis at pH 5.5. These results offer a convenient and traceless approach to protein and antibody functionalization.
Collapse
Affiliation(s)
- Daniele Melodia
- School of Chemistry, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Zachary Di Pietro
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Cheng Cao
- School of Chemistry, UNSW Sydney, Kensington, NSW 2052, Australia
| | | | - Robert Chapman
- School of Chemistry, UNSW Sydney, Kensington, NSW 2052, Australia.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
209
|
Xu Y, Zhu H, Li W, Chen D, Xu Y, Xu A, Ye D. Targeting adipokines in polycystic ovary syndrome and related metabolic disorders: from experimental insights to clinical studies. Pharmacol Ther 2022; 240:108284. [PMID: 36162728 DOI: 10.1016/j.pharmthera.2022.108284] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) affects approximately 15% of women of reproductive age worldwide. It is the most prevalent endocrine disorder with marked risks for female infertility, type 2 diabetes mellitus (T2DM), psychiatric disorders and gynecological cancers. Although the pathophysiology of PCOS remains largely elusive, growing evidence suggests a close link with obesity and its related metabolic disorders. As a highly active endocrine cell population, hypertrophic adipocytes in obesity have disturbed production of a vast array of adipokines, biologically active peptides that exert pleiotropic effects on homeostatic regulation of glucose and lipid metabolism. In parallel with their crucial roles in the pathophysiology of obesity-induced metabolic diseases, adipokines have recently been identified as promising targets for novel therapeutic strategies for multiple diseases. Current treatments for PCOS are suboptimal with insufficient alleviation of all symptoms. Novel findings in adipokine-targeted agents may provide important insight into the development of new drugs for PCOS. This Review presents an overview of the current understanding of mechanisms that link PCOS to obesity and highlights emerging evidence of adipose-ovary crosstalk as a pivotal mediator of PCOS pathogenesis. We summarize recent findings of preclinical and clinical studies that reveal the therapeutic potential of adipokine-targeted novel approaches to PCOS and its related metabolic disorders. We also discuss the critical gaps in knowledge that need to be addressed to guide the development of adipokine-based novel therapies for PCOS.
Collapse
Affiliation(s)
- Yidan Xu
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huiqiu Zhu
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Weiwei Li
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Danxia Chen
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Xu
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Dewei Ye
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China; Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Metabolic Phenotyping in Model Animals, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
210
|
Eltaher HM, Blokpoel Ferreras LA, Jalal AR, Dixon JE. Direct contact-mediated non-viral gene therapy using thermo-sensitive hydrogel-coated dressings. BIOMATERIALS ADVANCES 2022; 143:213177. [PMID: 36371970 DOI: 10.1016/j.bioadv.2022.213177] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Nanotechnologies are being increasingly applied as systems for peptide and nucleic acid macromolecule drug delivery. However systemic targeting of these, or efficient topical and localized delivery remains an issue. A controlled release system that can be patterned and locally administered such as topically to accessible tissue (skin, eye, intestine) would therefore be transformative in realizing the potential of such strategies. We previously developed a technology termed GAG-binding enhanced transduction (GET) to efficiently deliver a variety of cargoes intracellularly, using GAG-binding peptides to mediate cell targeting, and cell penetrating peptides (CPPs) to promote uptake. Herein we demonstrate that the GET transfection system can be used with the moisturizing thermo-reversible hydrogel Pluronic-F127 (PF127) and methyl cellulose (MC) to mediate site specific and effective intracellular transduction and gene delivery through GET nanoparticles (NPs). We investigated hydrogel formulation and the temperature dependence of delivery, optimizing the delivery system. GET-NPs retain their activity to enhance gene transfer within our formulations, with uptake transferred to cells in direct contact with the therapy-laden hydrogel. By using Azowipe™ material in a bandage approach, we were able to show for the first-time localized gene transfer in vitro on cell monolayers. The ability to simply control localization of gene delivery on millimetre scales using contact-mediated transfer from moisture-providing thermo-reversible hydrogels will facilitate new drug delivery methods. Importantly our technology to site-specifically deliver the activity of novel nanotechnologies and gene therapeutics could be transformative for future regenerative medicine.
Collapse
Affiliation(s)
- Hoda M Eltaher
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom, NG7 2RD, UK; Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt, 21521
| | - Lia A Blokpoel Ferreras
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom, NG7 2RD, UK
| | - Aveen R Jalal
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom, NG7 2RD, UK
| | - James E Dixon
- Division of Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom, NG7 2RD, UK.
| |
Collapse
|
211
|
Jagdale P, Sepp A, Shah DK. Physiologically-based pharmacokinetic model for pulmonary disposition of protein therapeutics in humans. J Pharmacokinet Pharmacodyn 2022; 49:607-624. [PMID: 36266517 PMCID: PMC9589728 DOI: 10.1007/s10928-022-09824-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/06/2022] [Indexed: 10/28/2022]
Abstract
Lung related disorders like COPD and Asthma, as well as various infectious diseases, form a major therapeutic area which would benefit from a predictive and adaptable mathematical model for describing pulmonary disposition of biological modalities. In this study we fill that gap by extending the cross-species two-pore physiologically-based pharmacokinetic (PBPK) platform with more detailed respiratory tract that includes the airways and alveolar space with epithelial lining fluid. We parameterize the paracellular and FcRn-facilitated exchange pathways between the epithelial lining fluid and lung interstitial space by building a mechanistic model for the exchange between the two. The optimized two-pore PBPK model described pulmonary exposure of several systemically dosed mAbs for which data is available and is also in agreement with the observed levels of endogenous IgG and albumin. The proposed framework can be used to assess pharmacokinetics of new lung-targeting biologic therapies and guide their dosing to achieve desired exposure at the pulmonary site-of-action.
Collapse
Affiliation(s)
- Prabhas Jagdale
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA
| | - Armin Sepp
- Simcyp Division, Certara UK Ltd, 1 Concourse Way, Level 2-Acero, Sheffield, S1 2BJ, UK
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA.
| |
Collapse
|
212
|
Bizeau J, Adam A, Nadal C, Francius G, Siniscalco D, Pauly M, Bégin-Colin S, Mertz D. Protein sustained release from isobutyramide-grafted stellate mesoporous silica nanoparticles. Int J Pharm X 2022; 4:100130. [PMID: 36156982 PMCID: PMC9494245 DOI: 10.1016/j.ijpx.2022.100130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 10/31/2022] Open
Abstract
Proteins are great therapeutic candidates as endogenous biomolecules providing a wide range of applications. However, their delivery suffers from some limitations and specifically designed delivery systems having an efficient protein anchoring and delivery strategy are still needed. In this work, we propose to combine large pore stellate mesoporous silica (STMS) with isobutyramide (IBAM), as a "glue" molecule which has been shown promising for immobilization of various biomacromolecules at silica surface. We address here for the first time the ability of such IBAM-modified NPs to sustainably deliver proteins over a prolonged time. In this work, a quantitative loading study of proteins (serum albumin (HSA), peroxidase (HRP), immunoglobulin (IgG) and polylysine (PLL)) on STMS@IBAM is first presented using three complementary detection techniques to ensure precision and avoid protein quantification issues. The results demonstrated a high loading capacity for HSA and HRP (≥ ca. 350 μg.mg-1) but a moderate one for IgG and PLL. After evaluating the physicochemical properties of the loaded particles and their stability over scaling-up and washings, the ability of STMS@IBAM to release proteins over prolonged time was evaluated in equilibrium (static) and flow mimicking (dynamic) conditions and at different temperatures (25, 37, 45 °C). Results show not only the potential of such "glue" functionalized STMS to release proteins in a sustained way, but also the retention of the biological activity of immobilized and released HRP, used as an enzyme model. Finally, an AFM-force spectroscopy study was conducted to decipher the interactions between IBAM and proteins, showing the involvement of different interactions in the adsorption and release processes.
Collapse
Affiliation(s)
- Joëlle Bizeau
- Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS), UMR-7504 CNRS-Université de Strasbourg, 23 rue du Lœss, BP 34 67034, Strasbourg, France
| | - Alexandre Adam
- Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS), UMR-7504 CNRS-Université de Strasbourg, 23 rue du Lœss, BP 34 67034, Strasbourg, France
| | - Clémence Nadal
- Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS), UMR-7504 CNRS-Université de Strasbourg, 23 rue du Lœss, BP 34 67034, Strasbourg, France
| | - Grégory Francius
- Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l'Environnement (LCPME), UMR 7564 CNRS-Université de Lorraine, 405 rue de Vandoeuvre, 54600 Villers-lès-Nancy, France
| | - David Siniscalco
- Laboratoire de Chimie Physique et Microbiologie pour les Matériaux et l'Environnement (LCPME), UMR 7564 CNRS-Université de Lorraine, 405 rue de Vandoeuvre, 54600 Villers-lès-Nancy, France
| | - Matthias Pauly
- Université de Strasbourg, CNRS, Institut Charles Sadron (UPR22), 23 rue du Loess, 67034, Strasbourg BP 84047, France
| | - Sylvie Bégin-Colin
- Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS), UMR-7504 CNRS-Université de Strasbourg, 23 rue du Lœss, BP 34 67034, Strasbourg, France
| | - Damien Mertz
- Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS), UMR-7504 CNRS-Université de Strasbourg, 23 rue du Lœss, BP 34 67034, Strasbourg, France
| |
Collapse
|
213
|
Emerging application of hydrocyclone in biotechnology and food processing. Sep Purif Technol 2022. [DOI: 10.1016/j.seppur.2022.122992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
214
|
Wright L, Barnes TJ, Joyce P, Prestidge CA. Optimisation of a High-Throughput Model for Mucus Permeation and Nanoparticle Discrimination Using Biosimilar Mucus. Pharmaceutics 2022; 14:2659. [PMID: 36559151 PMCID: PMC9782027 DOI: 10.3390/pharmaceutics14122659] [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: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
High-throughput permeation models are essential in drug development for timely screening of new drug and formulation candidates. Nevertheless, many current permeability assays fail to account for the presence of the gastrointestinal mucus layer. In this study, an optimised high-throughput mucus permeation model was developed employing a highly biorelevant mucus mimic. While mucus permeation is primarily conducted in a simple mucin solution, the complex chemistry, nanostructure and rheology of mucus is more accurately modelled by a synthetic biosimilar mucus (BSM) employing additional protein, lipid and rheology-modifying polymer components. Utilising BSM, equivalent permeation of various molecular weight fluorescein isothiocyanate-dextrans were observed, compared with native porcine jejunal mucus, confirming replication of the natural mucus permeation barrier. Furthermore, utilising synthetic BSM facilitated the analysis of free protein permeation which could not be quantified in native mucus due to concurrent proteolytic degradation. Additionally, BSM could differentiate between the permeation of poly (lactic-co-glycolic) acid nanoparticles (PLGA-NP) with varying surface chemistries (cationic, anionic and PEGylated), PEG coating density and size, which could not be achieved by a 5% mucin solution. This work confirms the importance of utilising highly biorelevant mucus mimics in permeation studies, and further development will provide an optimal method for high-throughput mucus permeation analysis.
Collapse
Affiliation(s)
| | | | | | - Clive A. Prestidge
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide 5000, Australia
| |
Collapse
|
215
|
Shirinichi F, Ibrahim T, Rodriguez M, Sun H. Assembling the best of two worlds: Biomolecule‐polymer nanoparticles via polymerization‐induced self‐assembly. JOURNAL OF POLYMER SCIENCE 2022. [DOI: 10.1002/pol.20220614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Farbod Shirinichi
- Department of Chemistry and Chemical & Biomedical Engineering, Tagliatela College of Engineering University of New Haven West Haven Connecticut USA
| | - Tarek Ibrahim
- Department of Chemistry and Chemical & Biomedical Engineering, Tagliatela College of Engineering University of New Haven West Haven Connecticut USA
| | - Mia Rodriguez
- Department of Chemistry and Chemical & Biomedical Engineering, Tagliatela College of Engineering University of New Haven West Haven Connecticut USA
| | - Hao Sun
- Department of Chemistry and Chemical & Biomedical Engineering, Tagliatela College of Engineering University of New Haven West Haven Connecticut USA
| |
Collapse
|
216
|
Yin SY, Hu Y, Zheng J, Li J, Yang R. Tannic Acid-Assisted Biomineralization Strategy for Encapsulation and Intracellular Delivery of Protein Drugs. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50583-50591. [PMID: 36322919 DOI: 10.1021/acsami.2c15205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Protein therapy has been considered to be one of the most direct and safe ways to regulate cell function and treat tumors. However, safe and effective intracellular delivery of protein drugs is still a key challenge. Herein, we developed a tannic acid-assisted biomineralization strategy for the encapsulation and intracellular delivery of protein drugs. RNase A and glucose oxidase (GOD) were choose as the protein drug model. RNase A, GOD, TA, and Mn2+ are mixed in one pot to attain RG@MT, and CaCO3 coating is subsequently carried out to construct RG@MT@C through biomineralization. Once RG@MT@C is endocytosed, the acidic environment of the lysosome will dissolve the protective layer of CaCO3 and produce plenty of CO2 to cause lysosome bursting, ensuring the lysosome escape of the RG@MT@C and thus releasing the generated TA-Mn2+, RNase A, and GOD into the cytoplasm. The released substances would activate starvation therapy, chemodynamic therapy, and protein therapy pathways to ensure a high performance of cancer therapy. Due to simple preparation, low toxicity, and controlled release in the tumor microenvironment, we expect it can realize efficient and nondestructive delivery of protein drugs and meet the needs for precise, high performance of synergistically antitumor therapy in biomedical applications.
Collapse
Affiliation(s)
- Sheng-Yan Yin
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yingcai Hu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jing Zheng
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jishan Li
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Ronghua Yang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
217
|
Elsayed Y, Kühl T, Dauer K, Sayin A, Wagner KG, Imhof D. Bioanalytical Workflow for Qualitative and Quantitative Assessment of Hot-Melt Extruded Lysozyme Formulations. ACS OMEGA 2022; 7:40836-40843. [PMID: 36406547 PMCID: PMC9670096 DOI: 10.1021/acsomega.2c03559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/07/2022] [Indexed: 06/16/2023]
Abstract
Structural and functional integrities of formulated proteins are key characteristics that provide a better understanding of influencing factors and their adjustment during formulation development. Here, the procedures commonly used for protein analysis were applied and optimized to obtain a higher degree of accuracy, reproducibility, and reliability for the analysis of lysozyme extracts from hot-melt extrudates (HME). The extrudates were prepared with polyethylene glycol 20 000. The test lysozyme HMEs were subjected to extraction procedures and analytical methods following the International Council of Harmonization guidelines for testing the active protein ingredient Q 1 A (R2) in its pure and formulated form. Therefore, reversed-phase high-pressure liquid chromatography, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, matrix-assisted laser desorption ionization mass spectrometry, and fluorescence-based activity measurements were applied to study lysozyme stability and function after formulation. Long-term accelerated stability studies were performed for the pure and formulated protein. Our findings revealed a high degree of stability for lysozyme toward different temperatures and storage times, confirming that HME is a suitable formulation alternative that preserves lysozyme's properties and stability. The presented methods and workflow are recommended to be exploited for further protein drugs to assess usability and compatibility concerning different pharmaceutical applications.
Collapse
Affiliation(s)
- Yomnah
Y. Elsayed
- Department
of Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
- Department
of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Cairo 11566, Egypt
| | - Toni Kühl
- Department
of Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Katharina Dauer
- Department
of Pharmaceutics, Pharmaceutical Institute, University of Bonn, Gerhard-Domagk-Str. 3, Bonn D-53121, Germany
| | - Alina Sayin
- Department
of Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| | - Karl G. Wagner
- Department
of Pharmaceutics, Pharmaceutical Institute, University of Bonn, Gerhard-Domagk-Str. 3, Bonn D-53121, Germany
| | - Diana Imhof
- Department
of Pharmaceutical Biochemistry and Bioanalytics, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, Bonn D-53121, Germany
| |
Collapse
|
218
|
Dowman LJ, Kulkarni SS, Alegre-Requena JV, Giltrap AM, Norman AR, Sharma A, Gallegos LC, Mackay AS, Welegedara AP, Watson EE, van Raad D, Niederacher G, Huhmann S, Proschogo N, Patel K, Larance M, Becker CFW, Mackay JP, Lakhwani G, Huber T, Paton RS, Payne RJ. Site-selective photocatalytic functionalization of peptides and proteins at selenocysteine. Nat Commun 2022; 13:6885. [PMID: 36371402 PMCID: PMC9653470 DOI: 10.1038/s41467-022-34530-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2022] Open
Abstract
The importance of modified peptides and proteins for applications in drug discovery, and for illuminating biological processes at the molecular level, is fueling a demand for efficient methods that facilitate the precise modification of these biomolecules. Herein, we describe the development of a photocatalytic method for the rapid and efficient dimerization and site-specific functionalization of peptide and protein diselenides. This methodology, dubbed the photocatalytic diselenide contraction, involves irradiation at 450 nm in the presence of an iridium photocatalyst and a phosphine and results in rapid and clean conversion of diselenides to reductively stable selenoethers. A mechanism for this photocatalytic transformation is proposed, which is supported by photoluminescence spectroscopy and density functional theory calculations. The utility of the photocatalytic diselenide contraction transformation is highlighted through the dimerization of selenopeptides, and by the generation of two families of protein conjugates via the site-selective modification of calmodulin containing the 21st amino acid selenocysteine, and the C-terminal modification of a ubiquitin diselenide.
Collapse
Affiliation(s)
- Luke J Dowman
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sameer S Kulkarni
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Juan V Alegre-Requena
- Department of Chemistry, Colorado State University, Fort Collins, CO, 80523-1872, USA
| | - Andrew M Giltrap
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Alexander R Norman
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Ashish Sharma
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence in Exciton Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Liliana C Gallegos
- Department of Chemistry, Colorado State University, Fort Collins, CO, 80523-1872, USA
| | - Angus S Mackay
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Adarshi P Welegedara
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Emma E Watson
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Damian van Raad
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Gerhard Niederacher
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Susanne Huhmann
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Nicholas Proschogo
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Karishma Patel
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Mark Larance
- Charles Perkins Centre and School of Medical Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Christian F W Becker
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Joel P Mackay
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Girish Lakhwani
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence in Exciton Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Thomas Huber
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Robert S Paton
- Department of Chemistry, Colorado State University, Fort Collins, CO, 80523-1872, USA
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia.
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
219
|
Babović M, Shliaha PV, Gibb S, Jensen ON. Effective Amino Acid Sequencing of Intact Filgrastim by Multimodal Mass Spectrometry with Topdownr. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2022; 33:2087-2093. [PMID: 36263452 DOI: 10.1021/jasms.2c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Therapeutic proteins, known as biologicals, are an important and growing class of drugs for treatment of a series of human ailments. Amino acid sequence variants of therapeutic proteins can affect their safety and efficacy. Top-down mass spectrometry is well suited for the sequence analysis of intact therapeutic proteins. Fine-tuning of tandem mass spectrometry (MS/MS) fragmentation conditions is essential for maximizing the amino acid sequence coverage but is often time-consuming. We used topdownr, an automated and integrated multimodal approach to systematically assess high mass accuracy MS/MS fragmentation parameters to characterize filgrastim, a 19 kDa recombinant human granulocyte colony-stimulating factor used in treating neutropenia. A total of 276 different MS/MS conditions were systematically tested, including the following parameters: protein charge state, HCD and CID collision energy, ETD reaction time, ETD supplemental activation, and UVPD activation time. Stringent and accurate evaluation and annotation of the MS/MS data was achieved by requiring a fragment ion mass error of 5 ppm, considering reproducible N- and C-terminal fragment ions only, and excluding internal fragment ion assignments. We report the first EThcD and UVPD MS/MS analysis of intact filgrastim, and these two techniques combined resulted in 98% amino acid sequence coverage. By combining all tested fragmentation modes, we obtained near-complete amino acid sequence coverage (99.4%) of intact filgrastim.
Collapse
Affiliation(s)
- Maša Babović
- Department of Biochemistry and Molecular Biology and VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, 5230 Odense, Denmark
| | - Pavel V Shliaha
- Department of Biochemistry and Molecular Biology and VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, 5230 Odense, Denmark
| | - Sebastian Gibb
- Department of Anesthesiology and Intensive Care, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Ole N Jensen
- Department of Biochemistry and Molecular Biology and VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, 5230 Odense, Denmark
| |
Collapse
|
220
|
Advancing Biologic Therapy for Refractory Autoimmune Hepatitis. Dig Dis Sci 2022; 67:4979-5005. [PMID: 35147819 DOI: 10.1007/s10620-021-07378-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/27/2021] [Indexed: 01/05/2023]
Abstract
Biologic agents may satisfy an unmet clinical need for treatment of refractory autoimmune hepatitis. The goals of this review are to present the types and results of biologic therapy for refractory autoimmune hepatitis, indicate opportunities to improve and expand biologic treatment, and encourage comparative clinical trials. English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. Rituximab (monoclonal antibodies against CD20 on B cells), infliximab (monoclonal antibodies against tumor necrosis factor-alpha), low-dose recombinant interleukin 2 (regulatory T cell promoter), and belimumab (monoclonal antibodies against B cell activating factor) have induced laboratory improvement in small cohorts with refractory autoimmune hepatitis. Ianalumab (monoclonal antibodies against the receptor for B cell activating factor) is in clinical trial. These agents target critical pathogenic pathways, but they may also have serious side effects. Blockade of the B cell activating factor or its receptors may disrupt pivotal B and T cell responses, and recombinant interleukin 2 complexed with certain interleukin 2 antibodies may selectively expand the regulatory T cell population. A proliferation-inducing ligand that enhances T cell proliferation and survival is an unevaluated, potentially pivotal, therapeutic target. Fully human antibodies, expanded target options, improved targeting precision, more effective delivery systems, and biosimilar agents promise to improve efficacy, safety, and accessibility. In conclusion, biologic agents target key pathogenic pathways in autoimmune hepatitis, and early experiences in refractory disease encourage clarification of the preferred target, rigorous clinical trial, and comparative evaluations.
Collapse
|
221
|
Tajoddin NN, Konermann L. Structural Dynamics of a Thermally Stressed Monoclonal Antibody Characterized by Temperature-Dependent H/D Exchange Mass Spectrometry. Anal Chem 2022; 94:15499-15509. [DOI: 10.1021/acs.analchem.2c03931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Nastaran N. Tajoddin
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Lars Konermann
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| |
Collapse
|
222
|
Meziadi A, Zuberi N, de Haan HW, Gauthier MA. Overcoming PEG─Protein Mutual Repulsion to Improve the Efficiency of PEGylation. Biomacromolecules 2022; 23:4948-4956. [DOI: 10.1021/acs.biomac.2c01192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ahlem Meziadi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes J3X 1S2, Quebec, Canada
| | - Navid Zuberi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes J3X 1S2, Quebec, Canada
| | - Hendrick W. de Haan
- Faculty of Science, University of Ontario Institute of Technology, Oshawa L1H 7K4, Ontario, Canada
| | - Marc A. Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes J3X 1S2, Quebec, Canada
| |
Collapse
|
223
|
Zhang S, Tan E, Wang R, Gao P, Wang H, Cheng Y. Robust Reversible Cross-Linking Strategy for Intracellular Protein Delivery with Excellent Serum Tolerance. NANO LETTERS 2022; 22:8233-8240. [PMID: 36173109 DOI: 10.1021/acs.nanolett.2c02948] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Intracellular protein delivery has attracted increasing attentions in biomedical applications. However, current delivery systems usually have poor serum stability due to the competitive binding of serum proteins to the polymers during delivery. Here, we report a reversible cross-linking strategy to improve the serum stability of polymers for robust intracellular protein delivery. In the proposed delivery system, nanoparticles are assembled by cargo proteins and cationic polymers and further stabilized by a glutathione-cleavable and traceless cross-linker. The cross-linked nanoparticles show high stability and efficient cell internalization in serum containing medium and can release the cargo proteins in response to intracellular glutathione and acidic pH in a traceless manner. The generality and versatility of the proposed strategy were demonstrated on different types of cationic polymers, cargo proteins, as well as cell lines. The study provides a facile and efficient method for improving the serum tolerance of cationic polymers in intracellular protein delivery.
Collapse
Affiliation(s)
- Song Zhang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Echuan Tan
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Ruijue Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Peng Gao
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
| | - Yiyun Cheng
- South China Advanced Institute for Soft Matter Science and Technology, School of Emergent Soft Matter, Guangdong Provincial Key Laboratory of Functional and Intelligent Hybrid Materials and Devices, South China University of Technology, Guangzhou, 510640, China
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, P.R. China
| |
Collapse
|
224
|
Church DC, Davis E, Caparco AA, Takiguchi L, Chung YH, Steinmetz NF, Pokorski JK. Polynorbornene-based bioconjugates by aqueous grafting-from ring-opening metathesis polymerization reduce protein immunogenicity. CELL REPORTS. PHYSICAL SCIENCE 2022; 3:101067. [PMID: 36816463 PMCID: PMC9933924 DOI: 10.1016/j.xcrp.2022.101067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Protein-polymer conjugates (PPCs) improve therapeutic efficacy of proteins and have been widely used for the treatment of various diseases such as cancer, diabetes, and hepatitis. PEGylation is considered as the "gold standard" in bioconjugation, although in practice its clinical applications are becoming limited because of extensive evidence of immunogenicity induced by pre-existing anti-PEG antibodies in patients. Here, optimized reaction conditions for living aqueous grafting-from ring-opening metathesis polymerization (ROMP) are utilized to synthesize water-soluble polynorbornene (PNB)-based PPCs of lysozyme (Lyz-PPCs) and bacteriophage Qβ (Qβ-PPCs) as PEG alternatives. Lyz-PPCs retain nearly 100% bioactivity and Qβ-PPCs exhibit up to 35% decrease in protein immunogenicity. Qβ-PPCs derived from NB-PEG show no reduction in recognition by anti-PEG antibodies while Qβ-PPCs derived from NB-Zwit show >95% reduction as compared with Qβ-PEG. This work demonstrates a new method for PPC synthesis and the utility of grafting from PPCs to evade immune recognition.
Collapse
Affiliation(s)
- Derek C. Church
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- These authors contributed equally
| | - Elizabathe Davis
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- These authors contributed equally
| | - Adam A. Caparco
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lauren Takiguchi
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Young Hun Chung
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jonathan K. Pokorski
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA 92093, USA
- Lead contact
| |
Collapse
|
225
|
Abstract
The homeostasis of cellular activities is essential for the normal functioning of living organisms. Hence, the ability to regulate the fates of cells is of great significance for both fundamental chemical biology studies and therapeutic development. Despite the notable success of small-molecule drugs that normally act on cellular protein functions, current clinical challenges have highlighted the use of macromolecules to tune cell function for improved therapeutic outcomes. As a class of hybrid biomacromolecules gaining rapidly increasing attention, protein conjugates have exhibited great potential as versatile tools to manipulate cell function for therapeutic applications, including cancer treatment, tissue engineering, and regenerative medicine. Therefore, recent progress in the design and assembly of protein conjugates used to regulate cell function is discussed in this review. The protein conjugates covered here are classified into three different categories based on their mechanisms of action and relevant applications: (1) regulation of intercellular interactions; (2) intervention in intracellular biological pathways; (3) termination of cell proliferation. Within each genre, a variety of protein conjugate scaffolds are discussed, which contain a diverse array of grafted molecules, such as lipids, oligonucleotides, synthetic polymers, and small molecules, with an emphasis on their conjugation methodologies and potential biomedical applications. While the current generation of protein conjugates is focused largely on delivery, the next generation is expected to address issues of site-specific conjugation, in vivo stability, controllability, target selectivity, and biocompatibility.
Collapse
Affiliation(s)
- Yiao Wang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Carston R Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mark D Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
226
|
Hatfield G, Tepliakova L, Gingras G, Stalker A, Li X, Aubin Y, Tam RY. Specific location of galactosylation in an afucosylated antiviral monoclonal antibody affects its FcγRIIIA binding affinity. Front Immunol 2022; 13:972168. [PMID: 36304448 PMCID: PMC9596277 DOI: 10.3389/fimmu.2022.972168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Monoclonal antibodies (mAbs) comprise an essential type of biologic therapeutics and are used to treat diseases because of their anti-cancer and anti-inflammatory properties, and their ability to protect against respiratory infections. Its production involves post-translational glycosylation, a biosynthetic process that conjugates glycans to proteins, which plays crucial roles in mAb bioactivities including effector functions and pharmacokinetics. These glycans are heterogeneous and have diverse chemical structures whose composition is sensitive to manufacturing conditions, rendering the understanding of how specific glycan structures affect mAb bioactivity challenging. There is a need to delineate the effects of specific glycans on mAb bioactivity to determine whether changes in certain glycosylation profiles (that can occur during manufacturing) will significantly affect product quality. Using enzymatic transglycosylation with chemically-defined N-glycans, we show that galactosylation at a specific location of N-glycans in an afucosylated anti-viral mAb is responsible for FcγRIIIA binding and antibody-dependent cell-mediated cytotoxicity (ADCC) activity. We report a facile method to obtain purified asymmetric mono-galactosylated biantennary complex N-glycans, and their influence on bioactivity upon incorporation into an afucosylated mAb. Using ELISA, surface plasmon resonance and flow cytometry, we show that galactosylation of the α6 antenna, but not the α3 antenna, consistently increases FcγRIIIA binding affinity. We confirm its relevance in an anti-viral model of respiratory syncytial virus (RSV) using an adapted ADCC reporter assay. We further correlate this structure-function relationship to the interaction of the galactose residue of the α6 antenna with the protein backbone using 2D-1H-15N-NMR, which showed that galactosylation of at this location exhibited chemical shift perturbations compared to glycoforms lacking this galactose residue. Our results highlight the importance of identifying and quantifying specific glycan isomers to ensure adequate quality control in batch-to-batch and biosimilar comparisons.
Collapse
|
227
|
Dou X, Xu Q, Dong B, Xu G, Qian N, Yang C, Li H, Chen L, Gao X, Song H. Anti-c-MET Fab-Grb2-Gab1 Fusion Protein-Mediated Interference of c-MET Signaling Pathway Induces Methuosis in Tumor Cells. Int J Mol Sci 2022; 23:ijms231912018. [PMID: 36233320 PMCID: PMC9569552 DOI: 10.3390/ijms231912018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Bio-macromolecules have potential applications in cancer treatment due to their high selectivity and efficiency in hitting therapeutic targets. However, poor cell membrane permeability has limited their broad-spectrum application in cancer treatment. The current study developed highly internalizable anti-c-MET antibody Fab fusion proteins with intracellular epitope peptide chimera to achieve the dual intervention from the extracellular to intracellular targets in tumor therapy. In vitro experiments demonstrated that the fusion proteins could interfere with the disease-associated intracellular signaling pathways and inhibit the uncontrolled proliferation of tumor cells. Importantly, investigation of the underlying mechanism revealed that these protein chimeras could induce vacuolation in treated cells, thus interfering with the normal extension and arrangement of microtubules as well as the mitosis, leading to the induction of methuosis-mediated cell death. Furthermore, in vivo tumor models indicated that certain doses of fusion proteins could inhibit the A549 xenograft tumors in NOD SCID mice. This study thus provides new ideas for the intracellular delivery of bio-macromolecules and the dual intervention against tumor cell signaling pathways.
Collapse
Affiliation(s)
- Xiaoqian Dou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Qinzhi Xu
- Beijng Immunoah Pharma Tech Co., Ltd., Beijing 100071, China
| | - Bo Dong
- Beijng Immunoah Pharma Tech Co., Ltd., Beijing 100071, China
| | - Guili Xu
- Beijng Immunoah Pharma Tech Co., Ltd., Beijing 100071, China
| | - Niliang Qian
- Beijng Immunoah Pharma Tech Co., Ltd., Beijing 100071, China
| | - Cuima Yang
- Beijng Immunoah Pharma Tech Co., Ltd., Beijing 100071, China
| | - Hongjie Li
- Beijng Immunoah Pharma Tech Co., Ltd., Beijing 100071, China
| | - Liting Chen
- Beijng Immunoah Pharma Tech Co., Ltd., Beijing 100071, China
| | - Xin Gao
- Beijng Immunoah Pharma Tech Co., Ltd., Beijing 100071, China
- Correspondence: (X.G.); (H.S.)
| | - Haifeng Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
- Correspondence: (X.G.); (H.S.)
| |
Collapse
|
228
|
Kumeria T, Wang J, Kim B, Park JH, Zuidema JM, Klempner M, Cavacini L, Wang Y, Sailor MJ. Enteric Polymer-Coated Porous Silicon Nanoparticles for Site-Specific Oral Delivery of IgA Antibody. ACS Biomater Sci Eng 2022; 8:4140-4152. [PMID: 36210772 PMCID: PMC10036216 DOI: 10.1021/acsbiomaterials.0c01313] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Porous silicon (pSi) nanoparticles are loaded with Immunoglobulin A-2 (IgA2) antibodies, and the assembly is coated with pH-responsive polymers on the basis of the Eudragit family of enteric polymers (L100, S100, and L30-D55). The temporal release of the protein from the nanocomposite formulations is quantified following an in vitro protocol simulating oral delivery: incubation in simulated gastric fluid (SGF; at pH 1.2) for 2 h, followed by a fasting state simulated intestinal fluid (FasSIF; at pH 6.8) or phosphate buffer solution (PBS; at pH 7.4). The nanocomposite formulations display a negligible release in SGF, while more than 50% of the loaded IgA2 is released in solutions at a pH of 6.8 (FasSIF) or 7.4 (PBS). Between 21 and 44% of the released IgA2 retains its functional activity. A capsule-based system is also evaluated, where the IgA2-loaded particles are packed into a gelatin capsule and the capsule is coated with either EudragitL100 or EudragitS100 polymer for a targeted release in the small intestine or the colon, respectively. The capsule-based formulations outperform polymer-coated nanoparticles in vitro, preserving 45-54% of the activity of the released protein.
Collapse
Affiliation(s)
- Tushar Kumeria
- Department of Chemistry and Biochemistry, University of California, San Diego, California 92093, United States
- School of Materials Science and Engineering, University of New South Wales-Sydney, Sydney, NSW 2052, Australia
| | - Joanna Wang
- Materials Science and Engineering Program, University of California, San Diego, California 92093, United States
| | - Byungji Kim
- Materials Science and Engineering Program, University of California, San Diego, California 92093, United States
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Korea
| | - Jonathan M Zuidema
- Department of Chemistry and Biochemistry, University of California, San Diego, California 92093, United States
| | - Mark Klempner
- MassBiologics of the University of Massachusetts Medical School, Boston, Massachusetts 02126, United States
| | - Lisa Cavacini
- MassBiologics of the University of Massachusetts Medical School, Boston, Massachusetts 02126, United States
| | - Yang Wang
- MassBiologics of the University of Massachusetts Medical School, Boston, Massachusetts 02126, United States
| | - Michael J Sailor
- Department of Chemistry and Biochemistry, University of California, San Diego, California 92093, United States
| |
Collapse
|
229
|
Kharandiuk T, Tan KH, Xu W, Weitenhagen F, Braun S, Göstl R, Pich A. Mechanoresponsive diselenide-crosslinked microgels with programmed ultrasound-triggered degradation and radical scavenging ability for protein protection. Chem Sci 2022; 13:11304-11311. [PMID: 36320583 PMCID: PMC9533411 DOI: 10.1039/d2sc03153a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
In the context of controlled delivery and release, proteins constitute a delicate class of cargo requiring advanced delivery platforms and protection. We here show that mechanoresponsive diselenide-crosslinked microgels undergo controlled ultrasound-triggered degradation in aqueous solution for the release of proteins. Simultaneously, the proteins are protected from chemical and conformational damage by the microgels, which disintegrate to water-soluble polymer chains upon sonication. The degradation process is controlled by the amount of diselenide crosslinks, the temperature, and the sonication amplitude. We demonstrate that the ultrasound-mediated cleavage of diselenide bonds in these microgels facilitates the release and activates latent functionality preventing the oxidation and denaturation of the encapsulated proteins (cytochrome C and myoglobin) opening new application possibilities in the targeted delivery of biomacromolecules.
Collapse
Affiliation(s)
- Tetiana Kharandiuk
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Worringerweg 1 52074 Aachen Germany
| | - Kok Hui Tan
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Worringerweg 1 52074 Aachen Germany
| | - Wenjing Xu
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Worringerweg 1 52074 Aachen Germany
| | - Fabian Weitenhagen
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Worringerweg 1 52074 Aachen Germany
| | - Susanne Braun
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Worringerweg 1 52074 Aachen Germany
| | - Robert Göstl
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany
| | - Andrij Pich
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Worringerweg 1 52074 Aachen Germany
- Aachen Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Brightlands Chemelot Campus Urmonderbaan 22, 6167 RD Geleen The Netherlands
| |
Collapse
|
230
|
Swinney DC. Why medicines work. Pharmacol Ther 2022; 238:108175. [DOI: 10.1016/j.pharmthera.2022.108175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 11/27/2022]
|
231
|
Kondengadan SM, Bansal S, Yang C, Liu D, Fultz Z, Wang B. Click chemistry and drug delivery: A bird’s-eye view. Acta Pharm Sin B 2022; 13:1990-2016. [DOI: 10.1016/j.apsb.2022.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/01/2022] Open
|
232
|
Lee SH, Yap WX, Jiang CQZ, Ler WX, Teo JWP, Ng SK, Heng D. Designing a ‘Ready-to-Use’ powder formulation platform for the inhaled protein therapeutics. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
233
|
Liu Y, Bai X, Lyu C, Fang J, Zhang F, Wu WH, Wei W, Zhang WB. Mechano-bioconjugation Strategy Empowering Fusion Protein Therapeutics with Aggregation Resistance, Prolonged Circulation, and Enhanced Antitumor Efficacy. J Am Chem Soc 2022; 144:18387-18396. [PMID: 36178288 DOI: 10.1021/jacs.2c06532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bioconjugation is a powerful protein modification strategy to improve protein properties. Herein, we report mechano-bioconjugation as a novel approach to empower fusion protein therapeutics and demonstrate its utility by a protein heterocatenane (cat-IFN-ABD) containing interferon-α2b (IFN) mechanically interlocked with a consensus albumin-binding domain (ABD). The conjugate was selectively synthesized in cellulo following a cascade of post-translational events using a pair of heterodimerizing p53dim variants and two orthogonal split-intein reactions. The catenane topology was proven by combined techniques of LC-MS, SDS-PAGE, SEC, and controlled proteolytic digestion. Not only did cat-IFN-ABD retain activities comparable to those of the wild-type IFN and ABD, the conjugate also exhibited enhanced aggregation resistance and prolonged circulation time over the simple linear and cyclic fusions. Consequently, cat-IFN-ABD potently inhibited tumor growth in the mouse xenograft model. Therefore, mechano-bioconjugation by catenation accomplishes function integration with additional benefits, providing an alternative pathway for developing advanced protein therapeutics.
Collapse
Affiliation(s)
- Yajie Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Center for Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| | - Xilin Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Center for Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| | - Chengliang Lyu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Jing Fang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Center for Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| | - Fan Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Center for Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| | - Wen-Hao Wu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Center for Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Wen-Bin Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Center for Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, P.R. China
| |
Collapse
|
234
|
Zhou W, Yang F, Li S, Yuan R, Xiang Y. A stimulus-responsive hexahedron DNA framework facilitates targeted and direct delivery of native anticancer proteins into cancer cells. Chem Sci 2022; 13:11132-11139. [PMID: 36320481 PMCID: PMC9516948 DOI: 10.1039/d2sc02858a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/08/2022] [Indexed: 11/30/2022] Open
Abstract
The targeted and direct intracellular delivery of proteins plays critical roles in biological research and disease treatments, yet remains highly challenging. Current solutions to such a challenge are limited by the modification of proteins that may potentially alter protein functions inside cells or the lack of targeting capability. Herein, we develop a stimulus-responsive and bivalent aptamer hexahedron DNA framework (HDF) for the targeted and direct delivery of native therapeutic proteins into cancer cells. The unmodified proteins are caged inside the HDF nanostructures assembled from six programmable single stranded DNAs to protect the proteins from degradation by cathepsins and enhance their targeting capability and delivery efficiency with the nanostructure-integrated aptamers. In addition, the protein drugs can be selectively released from the HDF nanostructures by the intracellular ATP molecules to induce tumor cell apoptosis, highlighting their promising application potential for cell biology and precise protein medicines.
Collapse
Affiliation(s)
- Wenjiao Zhou
- School of Chemistry and Chemical Engineering, Chongqing University of Technology Chongqing 400054 P. R. China
| | - Fang Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Shunmei Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| | - Yun Xiang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University Chongqing 400715 P. R. China
| |
Collapse
|
235
|
Pavel A, Saarimäki LA, Möbus L, Federico A, Serra A, Greco D. The potential of a data centred approach & knowledge graph data representation in chemical safety and drug design. Comput Struct Biotechnol J 2022; 20:4837-4849. [PMID: 36147662 PMCID: PMC9464643 DOI: 10.1016/j.csbj.2022.08.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/20/2022] Open
Abstract
Big Data pervades nearly all areas of life sciences, yet the analysis of large integrated data sets remains a major challenge. Moreover, the field of life sciences is highly fragmented and, consequently, so is its data, knowledge, and standards. This, in turn, makes integrated data analysis and knowledge gathering across sub-fields a demanding task. At the same time, the integration of various research angles and data types is crucial for modelling the complexity of organisms and biological processes in a holistic manner. This is especially valid in the context of drug development and chemical safety assessment where computational methods can provide solutions for the urgent need of fast, effective, and sustainable approaches. At the same time, such computational methods require the development of methodologies suitable for an integrated and data centred Big Data view. Here we discuss Knowledge Graphs (KG) as a solution to a data centred analysis approach for drug and chemical development and safety assessment. KGs are knowledge bases, data analysis engines, and knowledge discovery systems all in one, allowing them to be used from simple data retrieval, over meta-analysis to complex predictive and knowledge discovery systems. Therefore, KGs have immense potential to advance the data centred approach, the re-usability, and informativity of data. Furthermore, they can improve the power of analysis, and the complexity of modelled processes, all while providing knowledge in a natively human understandable network data model.
Collapse
Affiliation(s)
- Alisa Pavel
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Laura A Saarimäki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Lena Möbus
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,BioMediTech Institute, Tampere University, Tampere, Finland.,Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Tampere, Finland.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
236
|
Lee HJ, Choi ES, Maruoka K. Development of a catalytic ester activation protocol for the efficient formation of amide bonds using an Ar‐I/HF•pyridine/mCPBA system. ASIAN J ORG CHEM 2022. [DOI: 10.1002/ajoc.202200483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Hyo-Jun Lee
- Kunsan National University Department of Chemistry KOREA, REPUBLIC OF
| | - Eun-Sol Choi
- Kunsan National University Department of Chemistry KOREA, REPUBLIC OF
| | - Keiji Maruoka
- Kyoto University Graduate School of Pharmaceutical Sciences Sakyo 606-8501 Kyoto JAPAN
| |
Collapse
|
237
|
Cases
Díaz J, Lozano-Torres B, Giménez-Marqués M. Boosting Protein Encapsulation through Lewis-Acid-Mediated Metal-Organic Framework Mineralization: Toward Effective Intracellular Delivery. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2022; 34:7817-7827. [PMID: 36117882 PMCID: PMC9476658 DOI: 10.1021/acs.chemmater.2c01338] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/15/2022] [Indexed: 05/10/2023]
Abstract
Encapsulation of biomolecules using metal-organic frameworks (MOFs) to form stable biocomposites has been demonstrated to be a valuable strategy for their preservation and controlled release, which has been however restricted to specific electrostatic surface conditions. We present a Lewis-acid-mediated general in situ strategy that promotes the spontaneous MOF growth on a broad variety of proteins, for the first time, regardless of their surface nature. We demonstrate that MOFs based on cations exhibiting considerable inherent acidity such as MIL-100(Fe) enable efficient biomolecule encapsulation, including elusive alkaline proteins previously inaccessible by the well-developed in situ azolate-based MOF encapsulation. Specifically, we prove the MIL-100(Fe) scaffold for the encapsulation of a group of proteins exhibiting very different isoelectric points (5 < pI < 11), allowing triggered release under biocompatible conditions and retaining their activity after exposure to denaturing environments. Finally, we demonstrate the potential of the myoglobin-carrying biocomposite to facilitate the delivery of O2 into hypoxic human lung carcinoma A549 cells, overcoming hypoxia-associated chemoresistance.
Collapse
|
238
|
Reactive oxygen species-responsive branched poly (β-amino ester) with robust efficiency for cytosolic protein delivery. Acta Biomater 2022; 152:355-366. [PMID: 36084925 DOI: 10.1016/j.actbio.2022.08.070] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/29/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022]
Abstract
Protein therapy targeting the intracellular machinery holds great potentials for disease treatment, and therefore, effective cytosolic protein delivery technologies are highly demanded. Herein, we developed reactive oxygen species (ROS)-degradable, branched poly(β-amino ester) (PBAE) with built-in phenylboronic acid (PBA) in the backbone and terminal-pendent arginine for the efficient cytosolic protein delivery. The PBAE could form stable and cell-ingestible nanocomplexes (NCs) with proteins via electrostatic interaction, nitrogen-boronate (N-B) coordination, and hydrogen bonding, while it can be degraded into small segments by the over-produced H2O2 in tumor cells to enable cytoplasmic protein release. As thus, PBAE exhibited high efficiency in delivering varieties of proteins with distinct molecular weights (12.4-430 kDa) and isoelectric points (4.7-10.5) into tumor cells, including enzymes, toxins, and antibodies. Moreover, PBAE mediated efficient delivery of saporin into tumor cells in vivo, provoking pronounced anti-tumor outcomes. This study provides a robust and versatile platform for cytosolic protein delivery, and the elaborately tailored PBAE may find promising applications for protein-based biological research and disease management. STATEMENT OF SIGNIFICANCE: Cytosolic delivery of native proteins holds great therapeutic potentials, which however, is limited by the lack of robust delivery carriers that can simultaneously feature strong protein encapsulation yet effective intracellular protein release. Herein, ROS-degradable, branched poly(β-amino ester) (PBAE) with backbone-embedded phenylboronic acid (PBA) and terminal-pendent arginine was developed to synchronize these two processes. PBA and arginine moieties allowed PBAE to encapsulate proteins via N-B coordination, electrostatic interaction, hydrogen bonding, and salt bridging, while PBA could be oxidized by over-produced H2O2 inside cancer cells to trigger PBAE degradation and intracellular protein release. As thus, the top-performing PBAE mediated efficient cytosolic delivery of various proteins including enzymes, toxins, and antibodies. This study provides a powerful platform for cytosolic protein delivery, and may find promising utilities toward intracellular protein therapy against cancer and other diseases such as inflammation.
Collapse
|
239
|
Cheng Z, Li Y, Zhao D, Zhao W, Wu M, Zhang W, Cui Y, Zhang P, Zhang Z. Nanocarriers for intracellular co-delivery of proteins and small-molecule drugs for cancer therapy. Front Bioeng Biotechnol 2022; 10:994655. [PMID: 36147526 PMCID: PMC9485877 DOI: 10.3389/fbioe.2022.994655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
In the past few decades, the combination of proteins and small-molecule drugs has made tremendous progress in cancer treatment, but it is still not satisfactory. Because there are great differences in molecular weight, water solubility, stability, pharmacokinetics, biodistribution, and the ways of release and action between macromolecular proteins and small-molecule drugs. To improve the efficacy and safety of tumor treatment, people are committed to developing protein and drug co-delivery systems. Currently, intracellular co-delivery systems have been developed that integrate proteins and small-molecule drugs into one nanocarrier via various loading strategies. These systems significantly improve the blood stability, half-life, and biodistribution of proteins and small-molecule drugs, thus increasing their concentration in tumors. Furthermore, proteins and small-molecule drugs within these systems can be specifically targeted to tumor cells, and are released to perform functions after entering tumor cells simultaneously, resulting in improved effectiveness and safety of tumor treatment. This review summarizes the latest progress in protein and small-molecule drug intracellular co-delivery systems, with emphasis on the composition of nanocarriers, as well as on the loading methods of proteins and small-molecule drugs that play a role in cells into the systems, which have not been summarized by others so far.
Collapse
Affiliation(s)
- Zhihong Cheng
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Yongshuang Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Meng Wu
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Weilin Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yan Cui
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
240
|
Zhao Z, Liu X, Hou M, Zhou R, Wu F, Yan J, Li W, Zheng Y, Zhong Q, Chen Y, Yin L. Endocytosis-Independent and Cancer-Selective Cytosolic Protein Delivery via Reversible Tagging with LAT1 substrate. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110560. [PMID: 35789055 DOI: 10.1002/adma.202110560] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Protein drugs targeting intracellular machineries have shown profound therapeutic potentials, but their clinical utilities are greatly hampered by the lack of efficient cytosolic delivery techniques. Existing strategies mainly rely on nanocarriers or conjugated cell-penetrating peptides (CPPs), which often have drawbacks such as materials complexity/toxicity, lack of cell specificity, and endolysosomal entrapment. Herein, a unique carrier-free approach is reported for mediating cancer-selective and endocytosis-free cytosolic protein delivery. Proteins are sequentially modified with 4-nitrophenyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) benzyl carbonate as the H2 O2 -responsive domain and 3,4-dihydroxy-l-phenylalanine as the substrate of l-type amino acid transporter 1 (LAT1). Thus, the pro-protein can be directly transported into tumor cells by overexpressed LAT1 on cell membranes, bypassing endocytosis and endolysosomal entrapment. In the cytosol, overproduced H2 O2 restores the protein structure and activity. Using this technique, versatile proteins are delivered into tumor cells with robust efficiency, including toxins, enzymes, CRISPR-Cas9 ribonucleoprotein, and antibodies. Furthermore, intravenously injected pro-protein of saporin shows potent anticancer efficacy in 4T1-tumor-bearing mice, without provoking systemic toxicity. Such a facile and versatile pro-protein platform may benefit the development of protein pharmaceuticals.
Collapse
Affiliation(s)
- Ziyin Zhao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Xun Liu
- Department of Thoracic Surgery, Suzhou Key Laboratory of Thoracic Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Mengying Hou
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Renxiang Zhou
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Fan Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Jing Yan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Wei Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yujia Zheng
- Department of Thoracic Surgery, Suzhou Key Laboratory of Thoracic Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Qinmeng Zhong
- College of Chemistry, Chemical Engineering and Materials Science, Suzhou, 215123, China
| | - Yongbing Chen
- Department of Thoracic Surgery, Suzhou Key Laboratory of Thoracic Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Lichen Yin
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
241
|
An insight on lipid nanoparticles for therapeutic proteins delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
242
|
Yuan Z, Wan Z, Gao C, Wang Y, Huang J, Cai Q. Controlled magnesium ion delivery system for in situ bone tissue engineering. J Control Release 2022; 350:360-376. [PMID: 36002052 DOI: 10.1016/j.jconrel.2022.08.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
Abstract
Magnesium cation (Mg2+) has been an emerging therapeutic agent for inducing vascularized bone regeneration. However, the therapeutic effects of current magnesium (Mg) -containing biomaterials are controversial due to the concentration- and stage-dependent behavior of Mg2+. Here, we first provide an overview of biochemical mechanism of Mg2+ in various concentrations and suggest that 2-10 mM Mg2+in vitro may be optimized. This review systematically summarizes and discusses several types of controlled Mg2+ delivery systems based on polymer-Mg composite scaffolds and Mg-containing hydrogels, as well as their design philosophy and several parameters that regulate Mg2+ release. Given that the continuous supply of Mg2+ may prevent biomineral deposition in the later stage of bone regeneration and maturation, we highlight the controlled delivery of Mg2+ based dual- or multi-ions system, especially for the hierarchical therapeutic ion release system, which shows enhanced biomineralization. Finally, the remaining challenges and perspectives of Mg-containing biomaterials for future in situ bone tissue engineering are discussed as well.
Collapse
Affiliation(s)
- Zuoying Yuan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Zhuo Wan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Chenyuan Gao
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yue Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China.
| | - Qing Cai
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China..
| |
Collapse
|
243
|
The Effect of Trehalose Coating for Magnetite Nanoparticles on Stability of Egg White Lysozyme. Int J Mol Sci 2022; 23:ijms23179657. [PMID: 36077055 PMCID: PMC9456156 DOI: 10.3390/ijms23179657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 11/21/2022] Open
Abstract
In this study, the protein stability of hen egg-white lysozymes (HEWL) by Fe3O4 and Fe3O4-coated trehalose (Fe3O4@Tre) magnetic nanoparticles (NPs) is investigated. For this purpose, the co-precipitation method was used to synthesize magnetic NPs. The synthesized NPs were characterized by XRD, FT-IR spectroscopy, FE-SEM, and VSM analysis. In addition, the stability of HEWLs exposed to different NP concentrations in the range of 0.001–0.1 mg mL−1 was investigated by circular dichroism (CD) spectroscopy, fluorescence, and UV-Vis analysis. Based on the results, in the NP concentration range of 0.001–0.04 mg mL−1 the protein structure is more stable, and this range was identified as the range of kosmotropic concentration. The helicity was measured at two concentration points of 0.02 and 0.1 mg mL−1. According to the results, the α-helix at 0.02 mg mL−1 of Fe3O4 and Fe3O4@Tre was increased from 35.5% for native protein to 37.7% and 38.7%, respectively. The helicity decreased to 36.1% and 37.4%, respectively, with increasing the concentration of Fe3O4 and Fe3O4@Tre to 0.1 mg mL−1. The formation of hydrated water shells around protein molecules occurred by using Fe3O4@Tre NPs. Hence, it can be concluded that the trehalose as a functional group along with magnetic NPs can improve the stability of proteins in biological environments.
Collapse
|
244
|
Hwang SY, Park S, Jo H, Hee Seo S, Jeon KH, Kim S, Jung AR, Song C, Ahn M, Yeon Kwak S, Lee HJ, Uesugi M, Na Y, Kwon Y. Interrupting specific hydrogen bonds between ELF3 and MED23 as an alternative drug resistance-free strategy for HER2-overexpressing cancers. J Adv Res 2022; 47:173-187. [PMID: 35963541 PMCID: PMC10173165 DOI: 10.1016/j.jare.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION HER2 overexpression induces cancer aggression and frequent recurrences in many solid tumors. Because HER2 overproduction is generally followed by gene amplification, inhibition of protein-protein interaction (PPI) between transcriptional factor ELF3 and its coactivator MED23 has been considered an effective but challenging strategy. OBJECTIVES This study aimed to determine the hotspot of ELF3-MED23 PPI and further specify the essential residues and their key interactions in the hotspot which are controllable by small molecules with significant anticancer activity. METHODS Intensive biological evaluation methods including SEAP, fluorescence polarization, LC-MS/MS-based quantitative, biosensor, GST-pull down assays, and in silico structural analysis were performed to determine hotspot of ELF3-MED23 PPI and to elicit YK1, a novel small molecule PPI inhibitor. The effects of YK1 on possible PPIs of MED23 and the efficacy of trastuzumab were assessed using cell culture and tumor xenograft mouse models. RESULTS ELF3-MED23 PPI was found to be specifically dependent on H-bondings between D400, H449 of MED23 and W138, I140 of ELF3 for upregulating HER2 gene transcription. Employing YK1, we confirmed that interruption on these H-bondings significantly attenuated the HER2-mediated oncogenic signaling cascades and exhibited significant in vitro and in vivo anticancer activity against HER2-overexpressing breast and gastric cancers even in their trastuzumab refractory clones. CONCLUSION Our approach to develop specific ELF3-MED23 PPI inhibitor without interfering other PPIs of MED23 can finally lead to successful development of a drug resistance-free compound to interrogate HER2 biology in diverse conditions of cancers overexpressing HER2.
Collapse
Affiliation(s)
- Soo-Yeon Hwang
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Seojeong Park
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Hyunji Jo
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Seung Hee Seo
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Kyung-Hwa Jeon
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Seojeong Kim
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Ah-Reum Jung
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Chanju Song
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Misun Ahn
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Soo Yeon Kwak
- College of Pharmacy, CHA University, Pocheon 11160, Korea
| | - Hwa-Jong Lee
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Motonari Uesugi
- Institute for Chemical Research and Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Younghwa Na
- College of Pharmacy, CHA University, Pocheon 11160, Korea.
| | - Youngjoo Kwon
- College of Pharmacy & Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
245
|
Ko JH, Forsythe NL, Gelb MB, Messina KMM, Lau UY, Bhattacharya A, Olafsen T, Lee JT, Kelly KA, Maynard HD. Safety and Biodistribution Profile of Poly(styrenyl acetal trehalose) and Its Granulocyte Colony Stimulating Factor Conjugate. Biomacromolecules 2022; 23:3383-3395. [PMID: 35767465 DOI: 10.1021/acs.biomac.2c00511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Poly(styrenyl acetal trehalose) (pSAT), composed of trehalose side chains linked to a polystyrene backbone via acetals, stabilizes a variety of proteins and enzymes against fluctuations in temperature. A promising application of pSAT is conjugation of the polymer to therapeutic proteins to reduce renal clearance. To explore this possibility, the safety of the polymer was first studied. Investigation of acute toxicity of pSAT in mice showed that there were no adverse effects of the polymer at a high (10 mg/kg) concentration. The immune response (antipolymer antibody and cytokine production) in mice was also studied. No significant antipolymer IgG was detected for pSAT, and only a transient and low level of IgM was elicited. pSAT was also safe in terms of cytokine response. The polymer was then conjugated to a granulocyte colony stimulating factor (GCSF), a therapeutic protein that is approved by the Federal Drug Administration, in order to study the biodistribution of a pSAT conjugate. A site-selective, two-step synthesis approach was developed for efficient conjugate preparation for the biodistribution study resulting in 90% conjugation efficiency. The organ distribution of GCSF-pSAT was measured by positron emission tomography and compared to controls GCSF and GCSF-poly(ethylene glycol), which confirmed that the trehalose polymer conjugate improved the in vivo half-life of the protein by reducing renal clearance. These findings suggest that trehalose styrenyl polymers are promising for use in therapeutic protein-polymer conjugates for reduced renal clearance of the biomolecule.
Collapse
Affiliation(s)
- Jeong Hoon Ko
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Neil L Forsythe
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Madeline B Gelb
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Kathryn M M Messina
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Uland Y Lau
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Arvind Bhattacharya
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Tove Olafsen
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1569, United States
| | - Jason T Lee
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1569, United States
| | - Kathleen A Kelly
- Department of Pathology and Lab Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-1569, United States
| | - Heather D Maynard
- Department of Chemistry and Biochemistry and California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| |
Collapse
|
246
|
Itkonen J, Ghemtio L, Pellegrino D, Jokela (née Heinonen) PJ, Xhaard H, Casteleijn MG. Analysis of Biologics Molecular Descriptors towards Predictive Modelling for Protein Drug Development Using Time-Gated Raman Spectroscopy. Pharmaceutics 2022; 14:1639. [PMID: 36015265 PMCID: PMC9413954 DOI: 10.3390/pharmaceutics14081639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/29/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Pharmaceutical proteins, compared to small molecular weight drugs, are relatively fragile molecules, thus necessitating monitoring protein unfolding and aggregation during production and post-marketing. Currently, many analytical techniques take offline measurements, which cannot directly assess protein folding during production and unfolding during processing and storage. In addition, several orthogonal techniques are needed during production and market surveillance. In this study, we introduce the use of time-gated Raman spectroscopy to identify molecular descriptors of protein unfolding. Raman spectroscopy can measure the unfolding of proteins in-line and in real-time without labels. Using K-means clustering and PCA analysis, we could correlate local unfolding events with traditional analytical methods. This is the first step toward predictive modeling of unfolding events of proteins during production and storage.
Collapse
Affiliation(s)
- Jaakko Itkonen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00100 Helsinki, Finland
| | - Leo Ghemtio
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00100 Helsinki, Finland
| | - Daniela Pellegrino
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00100 Helsinki, Finland
| | - Pia J. Jokela (née Heinonen)
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00100 Helsinki, Finland
- Orion Pharma, 02101 Espoo, Finland
| | - Henri Xhaard
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00100 Helsinki, Finland
| | | |
Collapse
|
247
|
Negi S, Hamori M, Kawahara-Nakagawa Y, Imanishi M, Kurehara M, Kitada C, Kawahito Y, Kishi K, Manabe T, Kawamura N, Kitagishi H, Mashimo M, Shibata N, Sugiura Y. Importance of two-dimensional cation clusters induced by protein folding in intrinsic intracellular membrane permeability. RSC Chem Biol 2022; 3:1076-1084. [PMID: 35975000 PMCID: PMC9347356 DOI: 10.1039/d2cb00098a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
We investigated the cell penetration of Sp1 zinc finger proteins (Sp1 ZF) and the mechanism via which the total cationic charge and distribution of cationic residues on the protein surface affect intracellular trafficking. Sp1 ZFs showed intrinsic cell membrane permeability. The intracellular transfer of Sp1 ZFs other than 1F3 was dependent on the total cationic charge. Investigation of the effect of cationic residue distribution on intracellular membrane permeability revealed that the cellular uptake of unfolded Zn2+-non-coordinating Ala mutants was lower than that of the wild type. Therefore, the total cationic charge and distribution of cationic residues on the protein played crucial roles in intracellular translocation. Mutational studies revealed that the two-dimensional cation cluster on the protein surface significantly improved their cellular uptake. This study will contribute to the design of artificial cargoes that can efficiently transport target substances into cells.
Collapse
Affiliation(s)
- Shigeru Negi
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Mami Hamori
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Yuka Kawahara-Nakagawa
- Graduate School of Life Science, University of Hyogo 3-2-1 Kouto, Kamigori-cho Ako-gun Hyogo 678-1297 Japan
| | - Miki Imanishi
- Institute for Chemical Research, Kyoto University Uji Kyoto 611-0011 Japan
| | - Miku Kurehara
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Chieri Kitada
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Yuri Kawahito
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Kanae Kishi
- Graduate School of Biomedical and Health Sciences, Hiroshima University 1-2-3 Kasumi Minami-ku Hiroshima 734-8553 Japan
| | - Takayuki Manabe
- Clinical Research Support Center, Asahikawa Medical University Hospital 2-1-1-1 Midorigaokahigashi Asahikawa 078-8510 Japan
| | - Nobuyuki Kawamura
- Education Center for Pharmacy, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Applied Life Sciences 265-1 Higashijima, Akiha-ku Niigata City Niigata 956-8603 Japan
| | - Hiroaki Kitagishi
- Department of Molecular Chemistry and Biochemistry, Faculty of Science and Engineering, Doshisha University Kyotanabe Kyoto 610-0321 Japan
| | - Masato Mashimo
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Nobuhito Shibata
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| | - Yukio Sugiura
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo Kyotanabe Kyoto 610-0395 Japan
| |
Collapse
|
248
|
The Updated Review on Plant Peptides and Their Applications in Human Health. Int J Pept Res Ther 2022; 28:135. [PMID: 35911180 PMCID: PMC9326430 DOI: 10.1007/s10989-022-10437-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
Biologically active plant peptides, consisting of secondary metabolites, are compounds (amino acids) utilized by plants in their defense arsenal. Enzymatic processes and metabolic pathways secrete these plant peptides. They are also known for their medicinal value and have been incorporated in therapeutics of major human diseases. Nevertheless, its limitations (low bioavailability, high cytotoxicity, poor absorption, low abundance, improper metabolism, etc.) have demanded a need to explore further and discover other new plant compounds that overcome these limitations. Keeping this in mind, therapeutic plant proteins can be excellent remedial substitutes for bodily affliction. A multitude of these peptides demonstrates anti-carcinogenic, anti-microbial, anti-HIV, and neuro-regulating properties. This article's main aim is to list out and report the status of various therapeutic plant peptides and their prospective status as peptide-based drugs for multiple diseases (infectious and non-infectious). The feasibility of these compounds in the imminent future has also been discussed.
Collapse
|
249
|
Real-time imaging of monoclonal antibody film reconstitution after mechanical stress at the air-liquid interface by Brewster angle microscopy. Colloids Surf B Biointerfaces 2022; 218:112757. [DOI: 10.1016/j.colsurfb.2022.112757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022]
|
250
|
Controlled synthesis of PEGylated polyelectrolyte nanogels as efficient protein carriers. J Colloid Interface Sci 2022; 620:322-332. [DOI: 10.1016/j.jcis.2022.04.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/26/2022] [Accepted: 04/05/2022] [Indexed: 12/11/2022]
|