1
|
Liang X, Gupta D, Xie J, Van Wonterghem E, Van Hoecke L, Hean J, Niu Z, Ghaeidamini M, Wiklander OPB, Zheng W, Wiklander RJ, He R, Mamand DR, Bost J, Zhou G, Zhou H, Roudi S, Estupiñán HY, Rädler J, Zickler AM, Görgens A, Hou VWQ, Slovak R, Hagey DW, de Jong OG, Uy AG, Zong Y, Mäger I, Perez CM, Roberts TC, Carter D, Vader P, Esbjörner EK, de Fougerolles A, Wood MJA, Vandenbroucke RE, Nordin JZ, El Andaloussi S. Engineering of extracellular vesicles for efficient intracellular delivery of multimodal therapeutics including genome editors. Nat Commun 2025; 16:4028. [PMID: 40301355 PMCID: PMC12041237 DOI: 10.1038/s41467-025-59377-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
Intracellular delivery of protein and RNA therapeutics represents a major challenge. Here, we develop highly potent engineered extracellular vesicles (EVs) by incorporating bio-inspired attributes required for effective delivery. These comprise an engineered mini-intein protein with self-cleavage activity for active cargo loading and release, and fusogenic VSV-G protein for endosomal escape. Combining these components allows high efficiency recombination and genome editing in vitro following EV-mediated delivery of Cre recombinase and Cas9/sgRNA RNP cargoes, respectively. In vivo, infusion of a single dose Cre loaded EVs into the lateral ventricle in brain of Cre-LoxP R26-LSL-tdTomato reporter mice results in greater than 40% and 30% recombined cells in hippocampus and cortex respectively. In addition, we demonstrate therapeutic potential of this platform by showing inhibition of LPS-induced systemic inflammation via delivery of a super-repressor of NF-ĸB activity. Our data establish these engineered EVs as a platform for effective delivery of multimodal therapeutic cargoes, including for efficient genome editing.
Collapse
Affiliation(s)
- Xiuming Liang
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden.
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden.
- Cancer Research Laboratory, Shandong University-Karolinska Institutet collaborative Laboratory, School of Basic Medical Science, Shandong University, Jinan, Shandong, PR China.
| | - Dhanu Gupta
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford, OX3 7TY, UK
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
| | - Junhua Xie
- VIB Center for Inflammation Research, VIB, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, VIB, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Justin Hean
- Evox Therapeutics Limited, Oxford, United Kingdom
| | - Zheyu Niu
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Marziyeh Ghaeidamini
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, 41296, Göteborg, Sweden
| | - Oscar P B Wiklander
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Wenyi Zheng
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Rim Jawad Wiklander
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rui He
- Experimental Cancer Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet., Stockholm, Sweden
| | - Doste R Mamand
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Jeremy Bost
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Guannan Zhou
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, 419 Fang-Xie Road, Shanghai, 200011, P.R. China
| | - Houze Zhou
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Samantha Roudi
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - H Yesid Estupiñán
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
- Departamento de Ciencias Basicas, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Julia Rädler
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Antje M Zickler
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - André Görgens
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Vicky W Q Hou
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Radka Slovak
- Evox Therapeutics Limited, Oxford, United Kingdom
| | - Daniel W Hagey
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden
| | - Olivier G de Jong
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Aileen Geobee Uy
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Yuanyuan Zong
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China
| | - Imre Mäger
- Institute of Technology, University of Tartu, 50 411, Tartu, Estonia
| | - Carla Martin Perez
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford, OX3 7TY, UK
| | - Thomas C Roberts
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford, OX3 7TY, UK
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
- MDUK Oxford Neuromuscular Centre, Oxford, OX3 7TY, UK
| | - Dave Carter
- Evox Therapeutics Limited, Oxford, United Kingdom
| | - Pieter Vader
- CDL Research, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Elin K Esbjörner
- Division of Chemical Biology, Department of Life Sciences, Chalmers University of Technology, 41296, Göteborg, Sweden
| | | | - Matthew J A Wood
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford, OX3 7TY, UK
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK
- MDUK Oxford Neuromuscular Centre, Oxford, OX3 7TY, UK
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Joel Z Nordin
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden.
- Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Sweden.
| | - Samir El Andaloussi
- Division for Biomolecular and Cellular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
- Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden.
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
2
|
Liu B, Gong S, Qiu J, Ejaz W, Thayumanavan S. Synergistic Effects of Polycationic and Polyfluorinated Functionalities for Efficient Intracellular Protein Delivery. Biomacromolecules 2025; 26:2413-2420. [PMID: 40020198 DOI: 10.1021/acs.biomac.4c01795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Intracellular protein therapy is a promising strategy in biologics, including vaccine development, gene editing, and cancer therapeutics. However, protein-based drug delivery remains a significant challenge, particularly in penetrating cell barriers to reach intracellular targets. Inspired by transport adjuvants, we designed a series of polymeric vectors to achieve efficient functional protein trafficking with low cytotoxicity. With an adequate combination of guanidinium and fluorocarbon functionalities, a synergistic improvement of intracellular delivery is achieved in terms of both high intracellular transport and low cellular toxicity. The observed synergistic outcomes highlight new opportunities for delivery vehicle optimizations of intracellular biologics.
Collapse
|
3
|
Gong S, Liu B, Qiu J, Huang F, Thayumanavan S. Antibody-Directing Antibody Conjugates (ADACs) Enabled by Orthogonal Click Chemistry for Targeted Intracellular Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402874. [PMID: 39162119 PMCID: PMC11581923 DOI: 10.1002/smll.202402874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/03/2024] [Indexed: 08/21/2024]
Abstract
Using orthogonal click chemistries for efficient nanoscale self-assembly, a new antibody-directing antibody conjugate (ADAC) nanogel is generated. In this system, one of the antibodies is displayed on the nanogel surface to specifically recognize cell-surface epitopes while the other antibody is encapsulated inside the nanogel core. The system is programmed to release the latter antibody in its functional form in the cytosolic environment of a specific cell to engage intracellular targets. ADACs offer a potential solution to harness the advantages seen with antibody-drug conjugates (ADCs) to deliver therapeutic cargos to specific tissues, but with the added capability of carrying biologics as the cargo. In this manuscript, this potential is demonstrated through delivery of antibodies against intracellular targets in specific cells. This platform offers new avenues for precise therapeutic interventions and the potential to address previously "undruggable" cellular targets.
Collapse
Affiliation(s)
- Shuai Gong
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Bin Liu
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Jingyi Qiu
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Fangying Huang
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| |
Collapse
|
4
|
Dutta K, Zheng T, Hetrick EM. Comparative understanding of peroxide quantitation assays: a case study with peptide drug product degradation. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:4755-4764. [PMID: 38953302 DOI: 10.1039/d4ay00652f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Peroxide-mediated oxidation of drug molecules is a known challenge faced throughout the pharmaceutical development pathway-from early-stage stability studies to manufacturing processes. During the initial development stage, the major source of peroxide is the formulation excipients, whether they are pre-loaded or generated in situ due to slow degradation, and in the late phase, peroxides can be introduced during sanitization processes or generated via cavitation. In essence, a control strategy for peroxide mitigation often becomes a critical quality attribute for successful drug development. To this end, quantitation of peroxide is essential to monitor the peroxide level to ensure product quality and proposed shelf-life. However, methods for reliable and robust quantitation to detect trace levels of peroxide in a complex drug product matrix become increasingly challenging. This article discusses three high-throughput assays based on absorbance, fluorescence and chemiluminescence measurements to detect peroxide at a low level and compares the methods through validation studies in water. Selected methods have also been tested to understand the forced degradation of model peptide drug products with spiked hydrogen peroxide. Peptide degradation profiles and residual peroxide levels are presented to provide an understanding of the suitability of the quantitation methods and their performance.
Collapse
Affiliation(s)
- Kingshuk Dutta
- Bioproduct Research & Development, Lilly Technology Center-North, Indianapolis, IN 46221, USA.
| | - Tao Zheng
- Bioproduct Research & Development, Lilly Technology Center-North, Indianapolis, IN 46221, USA.
| | - Evan M Hetrick
- Bioproduct Research & Development, Lilly Technology Center-North, Indianapolis, IN 46221, USA.
| |
Collapse
|
5
|
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
6
|
Liu X, Zhao Z, Li W, Li Y, Yang Q, Liu N, Chen Y, Yin L. Engineering Nucleotidoproteins for Base-Pairing-Assisted Cytosolic Delivery and Genome Editing. Angew Chem Int Ed Engl 2023; 62:e202307664. [PMID: 37718311 DOI: 10.1002/anie.202307664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
Protein therapeutics targeting intracellular machineries hold profound potential for disease treatment, and hence robust cytosolic protein delivery technologies are imperatively demanded. Inspired by the super-negatively charged, nucleotide-enriched structure of nucleic acids, adenylated pro-proteins (A-proteins) with dramatically enhanced negative surface charges have been engineered for the first time via facile green synthesis. Then, thymidine-modified polyethyleneimine is developed, which exhibits strong electrostatic attraction, complementary base pairing, and hydrophobic interaction with A-proteins to form salt-resistant nanocomplexes with robust cytosolic delivery efficiencies. The acidic endolysosomal environment enables traceless restoration of the A-proteins and consequently promotes the intracellular release of the native proteins. This strategy shows high efficiency and universality for a variety of proteins with different molecular weights and isoelectric points in mammalian cells. Moreover, it enables highly efficient delivery of CRISPR-Cas9 ribonucleoproteins targeting fusion oncogene EWSR1-FLI1, leading to pronounced anti-tumor efficacy against Ewing sarcoma. This study provides a potent and versatile platform for cytosolic protein delivery and gene editing, and may benefit the development of protein pharmaceuticals.
Collapse
Affiliation(s)
- Xun Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 215123, Suzhou, Jiangsu, China
- Department of Thoracic Cancer, The Second Affiliated Hospital of Soochow University, 215123, Suzhou, Jiangsu, China
| | - Ziyin Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Wei Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Yajie Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Qiang Yang
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Ningyu Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Yongbing Chen
- Department of Thoracic Cancer, The Second Affiliated Hospital of Soochow University, 215123, Suzhou, Jiangsu, China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 215123, Suzhou, Jiangsu, China
| |
Collapse
|
7
|
Kaltbeitzel J, Wich PR. Protein-based Nanoparticles: From Drug Delivery to Imaging, Nanocatalysis and Protein Therapy. Angew Chem Int Ed Engl 2023; 62:e202216097. [PMID: 36917017 DOI: 10.1002/anie.202216097] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/16/2023]
Abstract
Proteins and enzymes are versatile biomaterials for a wide range of medical applications due to their high specificity for receptors and substrates, high degradability, low toxicity, and overall good biocompatibility. Protein nanoparticles are formed by the arrangement of several native or modified proteins into nanometer-sized assemblies. In this review, we will focus on artificial nanoparticle systems, where proteins are the main structural element and not just an encapsulated payload. While under natural conditions, only certain proteins form defined aggregates and nanoparticles, chemical modifications or a change in the physical environment can further extend the pool of available building blocks. This allows the assembly of many globular proteins and even enzymes. These advances in preparation methods led to the emergence of new generations of nanosystems that extend beyond transport vehicles to diverse applications, from multifunctional drug delivery to imaging, nanocatalysis and protein therapy.
Collapse
Affiliation(s)
- Jonas Kaltbeitzel
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Peter R Wich
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
8
|
Gavriel K, van Doeselaar DCA, Geers DWT, Neumann K. Click'n lock: rapid exchange between unsymmetric tetrazines and thiols for reversible, chemoselective functionalisation of biomolecules with on-demand bioorthogonal locking. RSC Chem Biol 2023; 4:685-691. [PMID: 37654505 PMCID: PMC10467616 DOI: 10.1039/d3cb00062a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
The late-stage functionalisation and diversification of complex structures including biomolecules is often achieved with the help of click chemistry. Besides employing irreversible click-like reactions, many synthetic applications benefit from reversible click reaction strategies, so called de-/trans-click approaches. Yet, the combination of both, reversible and irreversible click chemistry - while still respecting the stringent criteria of click transformations - remains so far elusive for modifications of biomolecular structures. Here, we report click'n lock as a concept that enables reversible click reactions and on-demand locking of chemical entities, thus switching from reversible to irreversible modifications of complex biomolecules. For this purpose, we employ the tetrazine-thiol exchange (TeTEx) reaction as a fully traceless click reaction with second order rate constants k2 higher than 2 M-1 s-1 within aqueous environments. Employing TeTEx as a reversible click reaction for the chemoselective modification of biomolecules is made possible by the use of 3,6-disubstituted 1,2,4,5-tetrazines bearing a single sulfide residue. The inherent reactivity of tetrazines towards inverse electron demand Diels-Alder (IEDDA) reactions allows to stabilize the clicked structure, switching from reversible to irreversible systems (click'n lock).
Collapse
Affiliation(s)
- Katerina Gavriel
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University Nijmegen Heyendaalseweg 135 6525 AJ Nijmegen The Netherlands
| | - Dustin C A van Doeselaar
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University Nijmegen Heyendaalseweg 135 6525 AJ Nijmegen The Netherlands
| | - Daniëlle W T Geers
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University Nijmegen Heyendaalseweg 135 6525 AJ Nijmegen The Netherlands
| | - Kevin Neumann
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University Nijmegen Heyendaalseweg 135 6525 AJ Nijmegen The Netherlands
| |
Collapse
|
9
|
Porello I, Cellesi F. Intracellular delivery of therapeutic proteins. New advancements and future directions. Front Bioeng Biotechnol 2023; 11:1211798. [PMID: 37304137 PMCID: PMC10247999 DOI: 10.3389/fbioe.2023.1211798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Achieving the full potential of therapeutic proteins to access and target intracellular receptors will have enormous benefits in advancing human health and fighting disease. Existing strategies for intracellular protein delivery, such as chemical modification and nanocarrier-based protein delivery approaches, have shown promise but with limited efficiency and safety concerns. The development of more effective and versatile delivery tools is crucial for the safe and effective use of protein drugs. Nanosystems that can trigger endocytosis and endosomal disruption, or directly deliver proteins into the cytosol, are essential for successful therapeutic effects. This article aims to provide a brief overview of the current methods for intracellular protein delivery to mammalian cells, highlighting current challenges, new developments, and future research opportunities.
Collapse
|
10
|
Ren L, Jiang L, Ren Q, Lv J, Zhu L, Cheng Y. A light-activated polymer with excellent serum tolerance for intracellular protein delivery. Chem Sci 2023; 14:2046-2053. [PMID: 36845943 PMCID: PMC9945510 DOI: 10.1039/d2sc05848k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/20/2023] [Indexed: 01/22/2023] Open
Abstract
The design of efficient materials for intracellular protein delivery has attracted great interest in recent years; however, most current materials for this purpose are limited by poor serum stability due to the early release of cargoes triggered by abundant serum proteins. Here, we propose a light-activated crosslinking (LAC) strategy to prepare efficient polymers with excellent serum tolerance for intracellular protein delivery. A cationic dendrimer engineered with photoactivatable O-nitrobenzene moieties co-assembles with cargo proteins via ionic interactions, followed by light activation to yield aldehyde groups on the dendrimer and the formation of imine bonds with cargo proteins. The light-activated complexes show high stability in buffer and serum solutions, but dis-assemble under low pH conditions. As a result, the polymer successfully delivers cargo proteins green fluorescent protein and β-galactosidase into cells with maintained bioactivity even in the presence of 50% serum. The LAC strategy proposed in this study provides a new insight to improve the serum stability of polymers for intracellular protein delivery.
Collapse
Affiliation(s)
- Lanfang Ren
- 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 China
| | - Li Jiang
- School of Biomedical Engineering, Shanghai Jiaotong University Shanghai 200240 China
| | - Qianyi Ren
- 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 China
| | - Jia Lv
- 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 China
| | - Linyong Zhu
- School of Biomedical Engineering, Shanghai Jiaotong University Shanghai 200240 China
| | - Yiyun Cheng
- 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 China
| |
Collapse
|
11
|
Zhou T, Yuan S, Qian P, Wu Y. Enzymes in Nanomedicine for Anti-tumor Therapy. Chem Res Chin Univ 2023. [DOI: 10.1007/s40242-023-2349-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
12
|
Swierczynski MJ, Ding Y, Ball ZT. Dual-Boronic Acid Reagents That Combine Dynamic and Covalent Bioconjugation. Bioconjug Chem 2022; 33:2307-2313. [PMID: 36445785 DOI: 10.1021/acs.bioconjchem.2c00508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Boronic acids and boronate esters find appreciable use in chemical biology. Molecules containing orthogonal boronic acid pairs can be utilized for sequential metal-catalyzed cross-couplings for facile preparation of complex bioconjugates including protein-protein conjugates. In this paper, we expand bis-boronic acid reagents for tandem covalent and dynamic bioconjugation. Sequential cross-coupling of 2-nitroarylboronic acid with cysteine residues and condensation of phenylboronic acid with salicylhydroxamic acids (SHA) readily afforded bioconjugates under physiological conditions with dual covalent and dynamic linkages. Both small molecule- and macromolecule-protein conjugates were amenable with this approach and reversible upon addition of excess unfunctionalized SHA or reactive oxygen species. These investigations provide new insights into the kinetic stability of SHA adducts.
Collapse
Affiliation(s)
- Michael J Swierczynski
- Bioscience Research Collaborative, Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Yuxuan Ding
- Bioscience Research Collaborative, Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Zachary T Ball
- Bioscience Research Collaborative, Department of Chemistry, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| |
Collapse
|
13
|
Li Y, Champion JA. Self-assembling nanocarriers from engineered proteins: Design, functionalization, and application for drug delivery. Adv Drug Deliv Rev 2022; 189:114462. [PMID: 35934126 DOI: 10.1016/j.addr.2022.114462] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/09/2022] [Accepted: 07/15/2022] [Indexed: 01/24/2023]
Abstract
Self-assembling proteins are valuable building blocks for constructing drug nanocarriers due to their self-assembly behavior, monodispersity, biocompatibility, and biodegradability. Genetic and chemical modifications allow for modular design of protein nanocarriers with effective drug encapsulation, targetability, stimuli responsiveness, and in vivo half-life. Protein nanocarriers have been developed to deliver various therapeutic molecules including small molecules, proteins, and nucleic acids with proven in vitro and in vivo efficacy. This article reviews recent advances in protein nanocarriers that are not derived from natural protein nanostructures, such as protein cages or virus like particles. The protein nanocarriers described here are self-assembled from rationally or de novo designed recombinant proteins, as well as recombinant proteins complexed with other biomolecules, presenting properties that are unique from those of natural protein carriers. Design, functionalization, and therapeutic application of protein nanocarriers will be discussed.
Collapse
Affiliation(s)
- Yirui Li
- BioEngineering Program, Georgia Institute of Technology, United States
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, United States; BioEngineering Program, Georgia Institute of Technology, United States.
| |
Collapse
|
14
|
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
|
15
|
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: 4.3] [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
|
16
|
Papadimitriou L, Theodorou A, Papageorgiou M, Voutyritsa E, Papagiannaki A, Velonia K, Ranella A. pH responsive biohybrid BSA-poly(DPA) nanoparticles for interlysosomal drug delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103591] [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]
|
17
|
Ren Q, Chen Q, Ren L, Cao C, Liu R, Cheng Y. Amphipathic poly-β-peptides for intracellular protein delivery. Chem Commun (Camb) 2022; 58:4320-4323. [PMID: 35293911 DOI: 10.1039/d2cc00453d] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of amphipathic poly-β-peptides are designed for intracellular protein delivery. The poly-β-peptides with higher molecular weight and hydrophobic contents exhibit higher protein loading and superior delivery efficiency. The lead material efficiently delivers proteins into cells with reserved bioactivity.
Collapse
Affiliation(s)
- Qianyi Ren
- 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.
| | - Qi Chen
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Lanfang Ren
- 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.
| | - Chuntao Cao
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Runhui Liu
- State Key Laboratory of Bioreactor Engineering, Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yiyun Cheng
- 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
|
18
|
Liu X, Zhao Z, Wu F, Chen Y, Yin L. Tailoring Hyperbranched Poly(β-amino ester) as a Robust and Universal Platform for Cytosolic Protein Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108116. [PMID: 34894367 DOI: 10.1002/adma.202108116] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/26/2021] [Indexed: 05/24/2023]
Abstract
Cytosolic protein delivery is a prerequisite for protein-based biotechnologies and therapeutics on intracellular targets. Polymers that can complex with proteins to form nano-assemblies represent one of the most important categories of materials, because of the ease of nano-fabrication, high protein loading efficiency, no need for purification, and maintenance of protein bioactivity. Stable protein encapsulation and efficient intracellular liberation are two critical yet opposite processes toward cytosolic delivery, and polymers that can resolve these two conflicting challenges are still lacking. Herein, hyperbranched poly(β-amino ester) (HPAE) with backbone-embedded phenylboronic acid (PBA) is developed to synchronize these two processes, wherein PBA enhanced protein encapsulation via nitrogen-boronate (N-B) coordination while triggered polymer degradation and protein release upon oxidation by H2 O2 in cancer cells. Upon optimization of the branching degree, charge density, and PBA distribution, the best-performing A2-B3-C2-S2 -P2 is identified, which mediates robust delivery of various native proteins/peptides with distinct molecular weights (1.6-430 kDa) and isoelectric points (4.1-10.3) into cancer cells, including enzymes, toxins, antibodies, and CRISPR-Cas9 ribonucleoproteins (RNPs). Moreover, A2-B3-C2-S2 -P2 mediates effective cytosolic delivery of saporin both in vitro and in vivo to provoke remarkable anti-tumor efficacy. Such a potent and universal platform holds transformative potentials for protein pharmaceuticals.
Collapse
Affiliation(s)
- Xun Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Ziyin Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Fan Wu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yongbing Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
19
|
Horn JM, Obermeyer AC. Genetic and Covalent Protein Modification Strategies to Facilitate Intracellular Delivery. Biomacromolecules 2021; 22:4883-4904. [PMID: 34855385 PMCID: PMC9310055 DOI: 10.1021/acs.biomac.1c00745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein-based therapeutics represent a rapidly growing segment of approved disease treatments. Successful intracellular delivery of proteins is an important precondition for expanded in vivo and in vitro applications of protein therapeutics. Direct modification of proteins and peptides for improved cytosolic translocation are a promising method of increasing delivery efficiency and expanding the viability of intracellular protein therapeutics. In this Review, we present recent advances in both synthetic and genetic protein modifications for intracellular delivery. Active endocytosis-based and passive internalization pathways are discussed, followed by a review of modification methods for improved cytosolic delivery. After establishing how proteins can be modified, general strategies for facilitating intracellular delivery, such as chemical supercharging or inclusion of cell-penetrating motifs, are covered. We then outline protein modifications that promote endosomal escape. We finally examine the delivery of two potential classes of therapeutic proteins, antibodies and associated antibody fragments, and gene editing proteins, such as cas9.
Collapse
|
20
|
|
21
|
Stevens CA, Kaur K, Klok HA. Self-assembly of protein-polymer conjugates for drug delivery. Adv Drug Deliv Rev 2021; 174:447-460. [PMID: 33984408 DOI: 10.1016/j.addr.2021.05.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/22/2021] [Accepted: 05/03/2021] [Indexed: 01/07/2023]
Abstract
Protein-polymer conjugates are a class of molecules that combine the stability of polymers with the diversity, specificity, and functionality of biomolecules. These bioconjugates can result in hybrid materials that display properties not found in their individual components and can be particularly relevant for drug delivery applications. Engineering amphiphilicity into these bioconjugate materials can lead to phase separation and the assembly of high-order structures. The assembly, termed self-assembly, of these hierarchical structures entails multiple levels of organization: at each level, new properties emerge, which are, in turn, influenced by lower levels. Here, we provide a critical review of protein-polymer conjugate self-assembly and how these materials can be used for therapeutic applications and drug delivery. In addition, we discuss central bioconjugate design questions and propose future perspectives for the field of protein-polymer conjugate self-assembly.
Collapse
Affiliation(s)
- Corey A Stevens
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland.
| | - Kuljeet Kaur
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingénierie Chimiques, Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne, Switzerland
| |
Collapse
|
22
|
Shieh P, Hill MR, Zhang W, Kristufek SL, Johnson JA. Clip Chemistry: Diverse (Bio)(macro)molecular and Material Function through Breaking Covalent Bonds. Chem Rev 2021; 121:7059-7121. [PMID: 33823111 DOI: 10.1021/acs.chemrev.0c01282] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the two decades since the introduction of the "click chemistry" concept, the toolbox of "click reactions" has continually expanded, enabling chemists, materials scientists, and biologists to rapidly and selectively build complexity for their applications of interest. Similarly, selective and efficient covalent bond breaking reactions have provided and will continue to provide transformative advances. Here, we review key examples and applications of efficient, selective covalent bond cleavage reactions, which we refer to herein as "clip reactions." The strategic application of clip reactions offers opportunities to tailor the compositions and structures of complex (bio)(macro)molecular systems with exquisite control. Working in concert, click chemistry and clip chemistry offer scientists and engineers powerful methods to address next-generation challenges across the chemical sciences.
Collapse
Affiliation(s)
- Peyton Shieh
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Megan R Hill
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Wenxu Zhang
- Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Samantha L Kristufek
- 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
|
23
|
Liu B, Singh K, Gong S, Canakci M, Osborne BA, Thayumanavan S. Protein–Antibody Conjugates (PACs): A Plug‐and‐Play Strategy for Covalent Conjugation and Targeted Intracellular Delivery of Pristine Proteins. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103106] [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)
- Bin Liu
- Department of Chemistry University of Massachusetts Amherst MA 01003 USA
| | - Khushboo Singh
- Department of Chemistry University of Massachusetts Amherst MA 01003 USA
- Center for Bioactive Delivery Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| | - Shuai Gong
- Department of Chemistry University of Massachusetts Amherst MA 01003 USA
- Center for Bioactive Delivery Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| | - Mine Canakci
- Molecular and Cellular Biology Program University of Massachusetts Amherst MA 01003 USA
- Center for Bioactive Delivery Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| | - Barbara A. Osborne
- Molecular and Cellular Biology Program University of Massachusetts Amherst MA 01003 USA
- Center for Bioactive Delivery Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
- Department of Veterinary and Animal Sciences University of Massachusetts Amherst MA 01003 USA
| | - S. Thayumanavan
- Department of Chemistry University of Massachusetts Amherst MA 01003 USA
- Molecular and Cellular Biology Program University of Massachusetts Amherst MA 01003 USA
- Center for Bioactive Delivery Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
24
|
Liu B, Singh K, Gong S, Canakci M, Osborne B, Thayumanavan S. Protein-Antibody Conjugates (PACs): A Plug-and-Play Strategy for Covalent Conjugation and Targeted Intracellular Delivery of Pristine Proteins. Angew Chem Int Ed Engl 2021; 60:12813-12818. [PMID: 33768625 PMCID: PMC8762996 DOI: 10.1002/anie.202103106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Indexed: 12/17/2022]
Abstract
We report here on protein-antibody conjugates (PACs) that are used for antibody-directed delivery of protein therapeutics to specific cells. PACs have the potential to judiciously combine the merits of two prolific therapeutic approaches-biologics and antibody-drug conjugates. We utilize spherical polymer brushes to construct PACs using the combination of two simple and efficient functionally orthogonal click chemistries. In addition to the synthesis and characterization of these nanoparticles, we demonstrate that PACs are indeed capable of specifically targeting cells based on the presence of target antigen on the cell surface to deliver proteins. The potentially broad adaptability of PACs opens up new opportunities for targeted biologics in therapeutics and sensing.
Collapse
Affiliation(s)
- Bin Liu
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Khushboo Singh
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Shuai Gong
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Mine Canakci
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Barbara Osborne
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
| |
Collapse
|
25
|
Qamar B, Solomon M, Marin A, Fuerst TR, Andrianov AK, Muro S. Intracellular Delivery of Active Proteins by Polyphosphazene Polymers. Pharmaceutics 2021; 13:249. [PMID: 33578893 PMCID: PMC7916676 DOI: 10.3390/pharmaceutics13020249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
Achieving intracellular delivery of protein therapeutics within cells remains a significant challenge. Although custom formulations are available for some protein therapeutics, the development of non-toxic delivery systems that can incorporate a variety of active protein cargo and maintain their stability, is a topic of great relevance. This study utilized ionic polyphosphazenes (PZ) that can assemble into supramolecular complexes through non-covalent interactions with different types of protein cargo. We tested a PEGylated graft copolymer (PZ-PEG) and a pyrrolidone containing linear derivative (PZ-PYR) for their ability to intracellularly deliver FITC-avidin, a model protein. In endothelial cells, PZ-PYR/protein exhibited both faster internalization and higher uptake levels than PZ-PEG/protein, while in cancer cells both polymers achieved similar uptake levels over time, although the internalization rate was slower for PZ-PYR/protein. Uptake was mediated by endocytosis through multiple mechanisms, PZ-PEG/avidin colocalized more profusely with endo-lysosomes, and PZ-PYR/avidin achieved greater cytosolic delivery. Consequently, a PZ-PYR-delivered anti-F-actin antibody was able to bind to cytosolic actin filaments without needing cell permeabilization. Similarly, a cell-impermeable Bax-BH3 peptide known to induce apoptosis, decreased cell viability when complexed with PZ-PYR, demonstrating endo-lysosomal escape. These biodegradable PZs were non-toxic to cells and represent a promising platform for drug delivery of protein therapeutics.
Collapse
Affiliation(s)
- Bareera Qamar
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA;
| | - Melani Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
| | - Alexander Marin
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
| | - Thomas R. Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
| | - Alexander K. Andrianov
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; (M.S.); (A.M.); (T.R.F.)
- Institute of Catalonia for Research and Advanced Studies, 08010 Barcelona, Spain
- Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| |
Collapse
|
26
|
Dutta K, Kanjilal P, Das R, Thayumanavan S. Synergistic Interplay of Covalent and Non-Covalent Interactions in Reactive Polymer Nanoassembly Facilitates Intracellular Delivery of Antibodies. Angew Chem Int Ed Engl 2021; 60:1821-1830. [PMID: 33034131 PMCID: PMC7855684 DOI: 10.1002/anie.202010412] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/23/2020] [Indexed: 01/29/2023]
Abstract
The primary impediments in developing large antibodies as drugs against intracellular targets involve their low transfection efficiency and suitable reversible encapsulation strategies for intracellular delivery with retention of biological activity. To address this, we outline an electrostatics-enhanced covalent self-assembly strategy to generate polymer-protein/antibody nanoassemblies. Through structure-activity studies, we down-select the best performing self-immolative pentafluorophenyl containing activated carbonate polymer for bioconjugation. With the help of an electrostatics-aided covalent self-assembly approach, we demonstrate efficient encapsulation of medium to large proteins (HRP, 44 kDa and β-gal, 465 kDa) and antibodies (ca. 150 kDa). The designed polymeric nanoassemblies are shown to successfully traffic functional antibodies (anti-NPC and anti-pAkt) to cytosol to elicit their bioactivity towards binding intracellular protein epitopes and inducing apoptosis.
Collapse
Affiliation(s)
| | | | - Ritam Das
- University of Massachusetts, Amherst, MA, 01003, USA
| | - Sankaran Thayumanavan
- Department of Chemistry, Molecular and Cellular Biology Program, and The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
27
|
Guo J, Liu Y, Zha J, Han H, Chen Y, Jia Z. Enhancing the peroxidase-mimicking activity of hemin by covalent immobilization in polymer nanogels. Polym Chem 2021. [DOI: 10.1039/d0py01465f] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A polymeric nanozyme that can closely mimic peroxidase is presented. The coordination between pendant hemins and primary amines together with the synergistic interactions between substrates and nanogels contribute to the enhanced catalytic activity.
Collapse
Affiliation(s)
- Jieyu Guo
- Shenzhen Key Laboratory of Environmental Chemistry and Ecological Remediation
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| | - Yuting Liu
- Shenzhen Key Laboratory of Environmental Chemistry and Ecological Remediation
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| | - Junqi Zha
- Shenzhen Key Laboratory of Environmental Chemistry and Ecological Remediation
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| | - Honghua Han
- Shenzhen Key Laboratory of Environmental Chemistry and Ecological Remediation
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| | - Yantao Chen
- Shenzhen Key Laboratory of Environmental Chemistry and Ecological Remediation
- College of Chemistry and Environmental Engineering
- Shenzhen University
- Shenzhen
- China
| | - Zhongfan Jia
- Institute for Nanoscale Science and Technology
- College of Science and Engineering
- Flinders University
- South Australia 5042
- Australia
| |
Collapse
|
28
|
Chatterjee S, Anslyn EV, Bandyopadhyay A. Boronic acid based dynamic click chemistry: recent advances and emergent applications. Chem Sci 2020; 12:1585-1599. [PMID: 34163920 PMCID: PMC8179052 DOI: 10.1039/d0sc05009a] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/27/2020] [Indexed: 11/28/2022] Open
Abstract
Recently, reversible click reactions have found numerous applications in chemical biology, supramolecular chemistry, and biomedical applications. Boronic acid (BA)-mediated cis-diol conjugation is one of the best-studied reactions among them. An excellent understanding of the chemical properties and biocompatibility of BA-based compounds has inspired the exploration of novel chemistries using boron to fuel emergent sciences. This topical review focuses on the recent progress of iminoboronate and salicylhydroxamic-boronate constituted reversible click chemistries in the past decade. We highlight the mechanism of reversible kinetics and its applications in chemical biology, medicinal chemistry, biomedical devices, and material chemistry. This article also emphasizes the fundamental reactivity of these two conjugate chemistries with assorted nucleophiles at variable pHs, which is of utmost importance to any stimuli-responsive biological and material chemistry explorations.
Collapse
Affiliation(s)
- Saurav Chatterjee
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar Punjab-781039 India
| | - Eric V Anslyn
- Department of Chemistry, University of Texas 1 University Station A1590 Austin Texas 78712 USA
| | - Anupam Bandyopadhyay
- Biomimetic Peptide Engineering Laboratory, Department of Chemistry, Indian Institute of Technology Ropar Punjab-781039 India
| |
Collapse
|
29
|
Lv J, Wang C, Li H, Li Z, Fan Q, Zhang Y, Li Y, Wang H, Cheng Y. Bifunctional and Bioreducible Dendrimer Bearing a Fluoroalkyl Tail for Efficient Protein Delivery Both In Vitro and In Vivo. NANO LETTERS 2020; 20:8600-8607. [PMID: 33155820 DOI: 10.1021/acs.nanolett.0c03287] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Rational design of stimuli-responsive polymers for cytosolic protein delivery with robust efficiency is of great importance but remains a challenging task. Here, we reported a bioreducible and amphiphilic dendrimer bearing a fluoroalkyl tail for this purpose. The fluorolipid was conjugated to the focal point of a cysteamine-cored polyamidoamine dendrimer via disulfide bond, while phenylboronic acid moieties were decorated on dendrimer surface for efficient protein binding. The yielding polymer showed high protein binding capability and complex stability and could efficiently release the cargo proteins in a glutathione-responsive manner. The designed polymer was effective in the delivery of various membrane-impermeable proteins into living cells with reserved bioactivities. In addition, the polymer efficiently delivered a toxin protein saporin into 4T1 breast cancer cells and inhibited the tumor growth in vivo after intravenous injection. The developed polymer in this study is a promising vector for the delivery of membrane-impermeable proteins to treat various diseases.
Collapse
Affiliation(s)
- Jia Lv
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Changping Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Hongru Li
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhan Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Qianqian Fan
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ying Zhang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hui Wang
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yiyun Cheng
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou 510640, China
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
30
|
Li D, Li X, Yang F, Yuan R, Xiang Y. Targeted Delivery of DNA Framework-Encapsulated Native Therapeutic Protein into Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2020; 12:54489-54496. [PMID: 33253532 DOI: 10.1021/acsami.0c17887] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A protein-based therapy is significantly challenged by the successful delivery of native proteins into the targeted cancer cells. We address this challenge here using an all-sealed divalent aptamer tetrahedral DNA framework (asdTDF) delivery platform, in which the protein drug is encapsulated inside the cavity of the framework stoichiometrically via a reversible chemical bond. The ligase-assisted seal of the nicks results in highly enhanced TDF stability of the against nuclease digestion to effectively protect the therapeutic protein from degradation. In addition, the divalent aptamer sequences incorporated into the framework favor it with a target-specific and efficient delivery capability. Importantly, upon being readily delivered into the targeted cancer cells, endogenous glutathione can trigger the release of the native therapeutic protein from the TDF in a traceless fashion by cleaving the reversible chemical bond, thereby leading to effective apoptosis of the specific cancer cells.
Collapse
Affiliation(s)
- Daxiu Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Xia Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Fang Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| | - Yun Xiang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, PR China
| |
Collapse
|
31
|
Dutta K, Kanjilal P, Das R, Thayumanavan S. Synergistic Interplay of Covalent and Non‐Covalent Interactions in Reactive Polymer Nanoassembly Facilitates Intracellular Delivery of Antibodies. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202010412] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | | | - Ritam Das
- University of Massachusetts Amherst MA 01003 USA
| | - Sankaran Thayumanavan
- Department of Chemistry, Molecular and Cellular Biology Program, and The Center for Bioactive Delivery-Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
32
|
Zhang P, Zhang Y, Ding X, Shen W, Li M, Wagner E, Xiao C, Chen X. A Multistage Cooperative Nanoplatform Enables Intracellular Co-Delivery of Proteins and Chemotherapeutics for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2000013. [PMID: 33035385 DOI: 10.1002/adma.202000013] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 06/18/2020] [Indexed: 06/11/2023]
Abstract
Combining intracellularly active proteins with chemotherapeutics represents a promising strategy for synergistic cancer therapy. However, the lack of nanocarrier systems for delivery into cancer cells and controlled intracellular release of both physicochemically very distinct cargos significantly impedes the biomedical translation of this combination strategy in cancer therapy. Here, a well-designed triblock copolymer, mPEG-b-PGCA-b-PGTA, is reported for application in a multistage cooperative drug delivery nanoplatform that accomplishes effective intracellular co-delivery of hydrophilic ribonuclease A (RNase A) and hydrophobic doxorubicin (DOX). RNase A bioreversibly modified with phenylboronic acid groups via a ROS-cleavable carbamate linker is incorporated into the triblock copolymer nanoparticles with high efficiency through a pH-reversible phenylboronic acid-catechol linkage. The reversible covalent conjugations between RNase A and the triblock copolymer endow the nanoparticles with high stability under normal physiological conditions. Upon cellular internalization, the cooperative release of DOX and RNase A from the triblock copolymer nanoparticles is triggered at multiple stages by endosomal acidic environment and subsequent DOX-enhanced intracellular ROS environment. This leads to enhanced synergistic anticancer effects as demonstrated both in vitro and in vivo. Given the versatility of dynamic covalent conjugations, this work provides a universal and stable platform for intracellular co-delivery of various combinations of proteins and chemotherapeutics.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xiaoya Ding
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Wei Shen
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Center for NanoScience (CeNS), Ludwig-Maximilians-University, Munich, 81377, Germany
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
33
|
Li X, Wei Y, Wu Y, Yin L. Hypoxia‐Induced Pro‐Protein Therapy Assisted by a Self‐Catalyzed Nanozymogen. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202004008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Xudong Li
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 China
| | - Yuansong Wei
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 China
| | - Yuchen Wu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 China
| | - Lichen Yin
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 China
| |
Collapse
|
34
|
Li X, Wei Y, Wu Y, Yin L. Hypoxia‐Induced Pro‐Protein Therapy Assisted by a Self‐Catalyzed Nanozymogen. Angew Chem Int Ed Engl 2020; 59:22544-22553. [DOI: 10.1002/anie.202004008] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/17/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Xudong Li
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 China
| | - Yuansong Wei
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 China
| | - Yuchen Wu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 China
| | - Lichen Yin
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices Collaborative Innovation Center of Suzhou Nano Science and Technology Soochow University Suzhou 215123 China
| |
Collapse
|
35
|
Goswami R, Jeon T, Nagaraj H, Zhai S, Rotello VM. Accessing Intracellular Targets through Nanocarrier-Mediated Cytosolic Protein Delivery. Trends Pharmacol Sci 2020; 41:743-754. [PMID: 32891429 PMCID: PMC7502523 DOI: 10.1016/j.tips.2020.08.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 12/19/2022]
Abstract
Protein-based therapeutics have unique therapeutic potential due to their specificity, potency, and low toxicity. The vast majority of intracellular applications of proteins require access to the cytosol. Direct entry to the cytosol is challenging due to the impermeability of the cell membrane to proteins. As a result, multiple strategies have focused on endocytic uptake of proteins. Endosomally entrapped cargo, however, can have very low escape efficiency, with protein degradation occurring in acidic endolysosomal compartments. In this review, we briefly discuss endosomal escape strategies and review the strategy of cell membrane fusion, a recent strategy for direct delivery of proteins into the cell cytoplasm.
Collapse
Affiliation(s)
- Ritabrita Goswami
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | - Taewon Jeon
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Harini Nagaraj
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | - Shumei Zhai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
36
|
Liu B, Wu R, Gong S, Xiao H, Thayumanavan S. In Situ Formation of Polymeric Nanoassemblies Using an Efficient Reversible Click Reaction. Angew Chem Int Ed Engl 2020; 59:15135-15140. [PMID: 32410309 PMCID: PMC7666047 DOI: 10.1002/anie.202004017] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/02/2020] [Indexed: 12/25/2022]
Abstract
Polymer-drug conjugates are promising as strategies for drug delivery, because of their high drug loading capacity and low premature release profile. However, the preparation of these conjugates is often tedious. In this paper, we report an efficient method for polymer-drug conjugates using an ultrafast and reversible click reaction in a post-polymerization functionalization strategy. The reaction is based on the rapid condensation of boronic acid functionalities with salicylhydroxamates. The polymer, bearing the latter functionality, has been designed such that the reaction with boronic acid bearing drugs induces an in situ self-assembly of the conjugates to form well-defined nanostructures. We show that this method is not only applicable for molecules with an intrinsic boronic acid group, but also for the other molecules that can be linked to aryl boronic acids through a self-immolative linker. The linker has been designed to cause traceless release of the attached drug molecules, the efficiency of which has been demonstrated through intracellular delivery.
Collapse
Affiliation(s)
- Bin Liu
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| | - Ruiling Wu
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| | - Shuai Gong
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA, 01003, USA
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, MA, 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
37
|
Addy PS, Shivrayan M, Cencer M, Zhuang J, Moore JS, Thayumanavan S. Polymer with Competing Depolymerization Pathways: Chain Unzipping versus Chain Scission. ACS Macro Lett 2020; 9:855-859. [PMID: 35648518 DOI: 10.1021/acsmacrolett.0c00250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Interest in triggered depolymerization is growing, driven by needs in sustainable plastics, self-healing materials, controlled release, and sensory amplification. For many triggered depolymerization reactions, the rate-limiting step does not directly involve the stimulus, and therefore, depolymerization kinetics exhibit only weak or no correlation to the concentration and reactivity of the stimulus. However, for many applications, a direct relationship between the stimulus and the depolymerization kinetics is desired. Here we designed, synthesized, and studied a polymer in which a nucleophile-induced chain scission (NICS) mechanism competes with the chain unzipping pathway. We find that the choice of the chain end functionality and the character of the nucleophile determines which of these is the predominant pathway. The NICS pathway was found to be dependent on the stimulus concentration, in contrast to the chain unzipping mechanism. We demonstrate transferability of these molecular-scale, structure-property relationships to nanoscale materials by formulating the polymers into host nanoparticles.
Collapse
|
38
|
Zhang N, Mei K, Guan P, Hu X, Zhao Y. Protein-Based Artificial Nanosystems in Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907256. [PMID: 32378796 DOI: 10.1002/smll.201907256] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 05/21/2023]
Abstract
Proteins, like actors, play different roles in specific applications. In the past decade, significant achievements have been made in protein-engineered biomedicine for cancer therapy. Certain proteins such as human serum albumin, working as carriers for drug/photosensitizer delivery, have entered clinical use due to their long half-life, biocompatibility, biodegradability, and inherent nonimmunogenicity. Proteins with catalytic abilities are promising as adjuvant agents for other therapeutic modalities or as anticancer drugs themselves. These catalytic proteins are usually defined as enzymes with high biological activity and substrate specificity. However, clinical applications of these kinds of proteins remain rare due to protease-induced denaturation and weak cellular permeability. Based on the characteristics of different proteins, tailor-made protein-based nanosystems could make up for their individual deficiencies. Therefore, elaborately designed protein-based nanosystems, where proteins serve as drug carriers, adjuvant agents, or therapeutic drugs to make full use of their intrinsic advantages in cancer therapy, are reviewed. Up-to-date progress on research in the field of protein-based nanomedicine is provided.
Collapse
Affiliation(s)
- Nan Zhang
- School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Kun Mei
- School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Ping Guan
- School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Xiaoling Hu
- School of Natural and Applied Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
39
|
Liu B, Ejaz W, Gong S, Kurbanov M, Canakci M, Anson F, Thayumanavan S. Engineered Interactions with Mesoporous Silica Facilitate Intracellular Delivery of Proteins and Gene Editing. NANO LETTERS 2020; 20:4014-4021. [PMID: 32298126 PMCID: PMC7351089 DOI: 10.1021/acs.nanolett.0c01387] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Intracellular delivery of functional proteins is a promising, but challenging, strategy for many therapeutic applications. Here, we report a new methodology that overcomes drawbacks of traditional mesoporous silica (MSi) particles for protein delivery. We hypothesize that engineering enhancement in interactions between proteins and delivery vehicles can facilitate efficient encapsulation and intracellular delivery. In this strategy, surface lysines in proteins were modified with a self-immolative linker containing a terminal boronic acid for stimulus-induced reversibility in functionalization. The boronic acid moiety serves to efficiently interact with amine-functionalized MSi through dative and electrostatic interactions. We show that proteins of different sizes and isoelectric points can be quantitatively encapsulated into MSi, even at low protein concentrations. We also show that the proteins can be efficiently delivered into cells with retention of activity. Utility of this approach is further demonstrated with gene editing in cells, through the delivery of a CRISPR/Cas9 complex.
Collapse
Affiliation(s)
- Bin Liu
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Wardah Ejaz
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Shuai Gong
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Myrat Kurbanov
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Mine Canakci
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Francesca Anson
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, USA
- Corresponding Author:
| |
Collapse
|
40
|
Lv J, Tan E, Wang Y, Fan Q, Yu J, Cheng Y. Tailoring guanidyl-rich polymers for efficient cytosolic protein delivery. J Control Release 2020; 320:412-420. [DOI: 10.1016/j.jconrel.2020.01.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/17/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022]
|
41
|
Ren L, Lv J, Wang H, Cheng Y. A Coordinative Dendrimer Achieves Excellent Efficiency in Cytosolic Protein and Peptide Delivery. Angew Chem Int Ed Engl 2020; 59:4711-4719. [DOI: 10.1002/anie.201914970] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Lanfang Ren
- Shanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal University Shanghai 200241 China
| | - Jia Lv
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| | - Hui Wang
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal University Shanghai 200241 China
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| |
Collapse
|
42
|
Ren L, Lv J, Wang H, Cheng Y. A Coordinative Dendrimer Achieves Excellent Efficiency in Cytosolic Protein and Peptide Delivery. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Lanfang Ren
- Shanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal University Shanghai 200241 China
| | - Jia Lv
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| | - Hui Wang
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal University Shanghai 200241 China
- South China Advanced Institute for Soft Matter Science and TechnologySchool of Molecular Science and EngineeringSouth China University of Technology Guangzhou 510640 China
| |
Collapse
|
43
|
Zhang S, Cheng Y. Boronic acid-engineered gold nanoparticles for cytosolic protein delivery. Biomater Sci 2020; 8:3741-3750. [DOI: 10.1039/d0bm00679c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Boronic acid-engineered gold nanoparticles for effective cytosolic protein delivery with the help of hypertonicity.
Collapse
Affiliation(s)
- Song Zhang
- South China Advanced Institute for Soft Matter Science and Technology
- School of Molecular Science and Engineering
- South China University of Technology
- Guangzhou 510640
- China
| | - Yiyun Cheng
- South China Advanced Institute for Soft Matter Science and Technology
- School of Molecular Science and Engineering
- South China University of Technology
- Guangzhou 510640
- China
| |
Collapse
|