1
|
Sun Y, Liu L, He H, Cui G, Zheng Y, Ye C, Qu L, Sun Y, Ji J, Lammers T, Zhang Y, Zhong Z. Co-activating STING-TLR9 pathways promotes radiotherapy-induced cancer vaccination. J Control Release 2025; 379:327-343. [PMID: 39778743 DOI: 10.1016/j.jconrel.2024.12.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Vaccination may cure cancer patients by inducing tumor-specific immune responses. Radiotherapy is an appealing strategy to generate cancer vaccines in situ; thus far, however, only modest and short-lived immune responses are achieved. We here show that radiation combined with co-activating STING-TLR9 can generate powerful in situ cancer vaccines. Notably, radiation at a dose of 12Gy is found to be optimal for boosting tumor cell immunogenicity, and STING-TLR9 co-stimulation by a dual immune activation nano-agonist overrides key immunosuppressive effects associated with radiotherapy. Local radiotherapy combined with the dual immune activation nano-agonists elicits strong systemic anti-tumor immune responses, resulting in complete regression of tumors and metastases in multiple syngeneic murine tumor models. This work introduces a novel and highly potent cancer immunotherapeutic strategy that holds promise for the personalized treatment of intractable cancers.
Collapse
Affiliation(s)
- Yu Sun
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Liang Liu
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; School of Optical and Electronic Information, Suzhou City University, Suzhou 215104, People's Republic of China
| | - Huilan He
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanhong Cui
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Yun Zheng
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Chunlian Ye
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Liping Qu
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Yinping Sun
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Jinlong Ji
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Clinic, Aachen 52074, Germany
| | - Ying Zhang
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China.
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
2
|
Sun H, Zhong Z. Bioresponsive Polymeric Nanoparticles: From Design, Targeted Therapy to Cancer Immunotherapy. Biomacromolecules 2025; 26:33-42. [PMID: 39667037 DOI: 10.1021/acs.biomac.4c01257] [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: 12/14/2024]
Abstract
Bioresponsive polymeric nanoparticles (NPs) that are capable of delivering and releasing therapeutics and biotherapeutics to target sites have attracted vivid interest in cancer therapy and immunotherapy. In contrast to enthusiastic evolution in the academic world, the clinical translation of these smart systems is scarce, partly due to concerns about safety, stability, complexity, and scalability. The moderate targetability, responsivity, and benefits are other concerns. In the past 17 years, we have devoted ourselves to exploring elegant strategies to address the above basic and translational problems by introducing diverse functional groups and/or targeting ligands to safe biomedical materials, such as biodegradable polymers and water-soluble polymers. This minimal modification is critical for further clinical translation. We have tailor-made various bioresponsive NPs including shell-sheddable and/or acid-sensitive biodegradable NPs, disulfide-cross-linked biodegradable micelles and polymersomes, and blood-brain barrier (BBB)-permeable NPs, to target different tumors. This perspective provides an overview of our work path toward targeted nanomedicines and personalized vaccines, which might inspire clinical translation and future research on cancer therapy.
Collapse
Affiliation(s)
- Huanli Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, 215222, PR China
| |
Collapse
|
3
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [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: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
4
|
Ahmad A, Khan JM, Paray BA, Rashid K, Parvez A. Endolysosomal trapping of therapeutics and endosomal escape strategies. Drug Discov Today 2024; 29:104070. [PMID: 38942071 DOI: 10.1016/j.drudis.2024.104070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/31/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
Internalizing therapeutic molecules or genes into cells and safely delivering them to the target tissue where they can perform the intended tasks is one of the key characteristics of the smart gene/drug delivery vector. Despite much research in this field, endosomal escape continues to be a significant obstacle to the development of effective gene/drug delivery systems. In this review, we discuss in depth the several types of endocytic pathways involved in the endolysosomal trapping of therapeutic agents. In addition, we describe numerous mechanisms involved in nanoparticle endosomal escape. Furthermore, many other techniques are employed to increase endosomal escape to minimize entrapment of therapeutic compounds within endolysosomes, which have been reviewed at length in this study.
Collapse
Affiliation(s)
- Aqeel Ahmad
- Department of Medical Biochemistry, College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia.
| | - Javed Masood Khan
- Department of Food Science and Nutrition, Faculty of Food and Agricultural Sciences, King Saud University, 2460, Riyadh 11451, Saudi Arabia
| | - Bilal Ahamad Paray
- Department of Zoology, College of Science, King Saud University, PO Box 2455, Riyadh 11451, Saudi Arabia
| | - Khalid Rashid
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ashib Parvez
- Department of Community Medicine, F.H. Medical College, Atal Bihari Vajpayee Medical University, Etmadpur, Agra, India
| |
Collapse
|
5
|
Ni D, Guo B, Zhong Z, Chen Y, Yang G, Yang J, Zhong Z, Meng F. Integrin-targeting disulfide-crosslinked micellar docetaxel eradicates lung and prostate cancer patient-derived xenografts. Acta Biomater 2023; 170:228-239. [PMID: 37634830 DOI: 10.1016/j.actbio.2023.08.043] [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: 05/27/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Actively targeted nanomedicines though conceptually attractive for tumor therapy are extremely hard to realize due to problems of premature drug leakage, excessive liver accretion, inadequate tumor uptake, and/or retarded drug release inside tumor cells. Here, we systemically studied the influence of disulfide crosslinking on the in vitro and in vivo performance of integrin-targeting micellar docetaxel (t-MDTX). Of note, t-M5DTX with a high disulfide content was clearly advantageous in terms of stability, intracellular drug release, anti-tumor activity toward αVβ3-overexpressing A549 cells, blood circulation and therapeutic efficacy in orthotopic A549-luc lung tumor-bearing mice. t-MDTX induced extraordinary tumor targetability with tumor-to-normal tissue ratios of 1.7-8.3. Further studies indicated that t-M5DTX could effectively eradicate αVβ3-overexpressing lung and prostate cancer patient-derived xenografts (PDX), in which ca. 80% mice became tumor-free. This integrin-targeting disulfide-crosslinked micellar docetaxel emerges as a promising actively targeted nanoformulation for tumor therapy. STATEMENT OF SIGNIFICANCE: Nanomedicines have a great potential in treating advanced tumor patients; however, their tumor-targeting ability and therapeutic efficacy remain unsatisfactory. In addition to PEGylation and ligand selection, particle size, stability and drug release behavior are also critical to their performance in vivo. In this paper, we find that small and cRGD-guided disulfide-crosslinked micellar docetaxel (t-MDTX) induces superior tumor uptake and retention but without increasing liver burden, leading to extraordinary selectivity and inhibition of αvβ3 overexpressing lung tumors. t-MDTX is further shown to effectively treat αvβ3-positive patient-derived tumor models, lending it a high potential for clinical translation.
Collapse
Affiliation(s)
- Dawei Ni
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Beibei Guo
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China
| | | | - Yu Chen
- GenePharma, Suzhou 215123, PR China
| | - Guang Yang
- Department of Oncology, Suzhou BenQ Medical Center, the Affiliated BenQ Hospital of Nanjing Medical University, Suzhou 215009, PR China
| | - Jiangtao Yang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China.
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
6
|
Lv W, Wu H, Zhang Y, Li H, Shu H, Su C, Zhu Y, Wang T, Nie F. cRGD-targeted gold-based nanoparticles overcome EGFR-TKI resistance of NSCLC via low-temperature photothermal therapy combined with sonodynamic therapy. Biomater Sci 2023; 11:1677-1691. [PMID: 36625328 DOI: 10.1039/d2bm01825j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) is a first-line targeted drug for the treatment of advanced non-small cell lung cancer (NSCLC) in clinical practice, but EGFR-TKI-acquired resistance limits its therapeutic effect. To address this challenge, a novel multifunctional gold-based targeted nanoparticle-based drug delivery system is fabricated. The gold-based nanoparticle is loaded with the EGFR-TKI (gefitinib) and IR780, and the surface-modified gold nanoshell layer has a photothermal effect for thermally triggered drug release. Finally, the unique binding of cyclic arginine-glycine-aspartic acid (cRGD) to the αvβ3 receptor ensured that the nanoparticle (cRGD-GIPG) targeted transport into drug-resistant NSCLC cells was functional. Due to the sonodynamic properties of IR780, ultrasound (US) irradiation promoted reactive oxygen species (ROS) generation, while low-temperature photothermal therapy (PTT) not only promoted the release of drug, but also further enhanced the cytotoxic effects of ROS. In turn, it blocked the activation of TGF-β/PDLIM5/SMAD resistance pathway and induced apoptosis of drug-resistant cells through mitochondrial apoptosis, enabling the treatment of EGFR-TKI-resistant NSCLC. The low-temperature PTT combined with sonodynamic therapy (SDT) by cRGD-GIPG thus shows potent anticancer activity against EGFR-TKI-resistant NSCLC cells in vitro and in vivo. The present work provides a valuable strategy for highly targeted and EGFR-TKI-resistant reversal therapy in NSCLC.
Collapse
Affiliation(s)
- Wenhao Lv
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, 730000, China. .,Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, 730000, China
| | - Hao Wu
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, 730000, China. .,Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, 730000, China
| | - Yao Zhang
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| | - Hui Li
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| | - Hong Shu
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| | - Chunhong Su
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| | - Yangyang Zhu
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, 730000, China. .,Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, 730000, China
| | - Ting Wang
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, 730000, China. .,Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, 730000, China
| | - Fang Nie
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, 730000, China. .,Gansu Province Clinical Research Center for Ultrasonography, Lanzhou, 730000, China
| |
Collapse
|
7
|
Gouveia MG, Wesseler JP, Ramaekers J, Weder C, Scholten PBV, Bruns N. Polymersome-based protein drug delivery - quo vadis? Chem Soc Rev 2023; 52:728-778. [PMID: 36537575 PMCID: PMC9890519 DOI: 10.1039/d2cs00106c] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Indexed: 12/24/2022]
Abstract
Protein-based therapeutics are an attractive alternative to established therapeutic approaches and represent one of the fastest growing families of drugs. While many of these proteins can be delivered using established formulations, the intrinsic sensitivity of proteins to denaturation sometimes calls for a protective carrier to allow administration. Historically, lipid-based self-assembled structures, notably liposomes, have performed this function. After the discovery of polymersome-based targeted drug-delivery systems, which offer manifold advantages over lipid-based structures, the scientific community expected that such systems would take the therapeutic world by storm. However, no polymersome formulations have been commercialised. In this review article, we discuss key obstacles for the sluggish translation of polymersome-based protein nanocarriers into approved pharmaceuticals, which include limitations imparted by the use of non-degradable polymers, the intricacies of polymersome production methods, and the complexity of the in vivo journey of polymersomes across various biological barriers. Considering this complex subject from a polymer chemist's point of view, we highlight key areas that are worthy to explore in order to advance polymersomes to a level at which clinical trials become worthwhile and translation into pharmaceutical and nanomedical applications is realistic.
Collapse
Affiliation(s)
- Micael G Gouveia
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Justus P Wesseler
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Jobbe Ramaekers
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Christoph Weder
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Philip B V Scholten
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Nico Bruns
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, UK
- Department of Chemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany.
| |
Collapse
|
8
|
Fabrication of Polymersomes: A Macromolecular Architecture in Nanotherapeutics. CHEMISTRY 2022. [DOI: 10.3390/chemistry4030070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In consideration of the issues of drug delivery systems, the artificial vesicle structures composed of block copolymers called polymersomes recently gained considerable attention. The possibility of tuning the mechanical parameter and increasing the scale-up production of polymersomes led to its wide application in healthcare. Bearing in mind the disease condition, the structure and properties of the polymersomes could be tuned to serve the purpose. Furthermore, specific ligands can be incorporated on the vesicular surface to induce smart polymersomes, thus improving targeted delivery. The synthesis method and surface functionalization are the two key aspects that determine the versatility of biological applications as they account for stability, specific targeting, degradability, biocompatibility, and bioavailability. A perfectly aligned polymer vesicle can mimic the cells/organelles and function by avoiding cytotoxicity. This supramolecular structure can carry and deliver payloads of a wide range, including drugs, proteins, and genes, contributing to the construction of next-generation therapeutics. These aspects promote the potential use of such components as a framework to approach damaged tissue while maintaining healthy environments during circulation. Herein, this article concentrates specifically on the drug delivery applications of polymersomes.
Collapse
|
9
|
Tian Y, Tirrell MV, LaBelle JL. Harnessing the Therapeutic Potential of Biomacromolecules through Intracellular Delivery of Nucleic Acids, Peptides, and Proteins. Adv Healthc Mater 2022; 11:e2102600. [PMID: 35285167 PMCID: PMC9232950 DOI: 10.1002/adhm.202102600] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/09/2022] [Indexed: 12/19/2022]
Abstract
Biomacromolecules have long been at the leading edge of academic and pharmaceutical drug development and clinical translation. With the clinical advances of new therapeutics, such as monoclonal antibodies and nucleic acids, the array of medical applications of biomacromolecules has broadened considerably. A major on-going effort is to expand therapeutic targets within intracellular locations. Owing to their large sizes, abundant charges, and hydrogen-bond donors and acceptors, advanced delivery technologies are required to deliver biomacromolecules effectively inside cells. In this review, strategies used for the intracellular delivery of three major forms of biomacromolecules: nucleic acids, proteins, and peptides, are highlighted. An emphasis is placed on synthetic delivery approaches and the major hurdles needed to be overcome for their ultimate clinical translation.
Collapse
Affiliation(s)
- Yu Tian
- Pritzker School of Molecular EngineeringThe University of Chicago5640 S Ellis AveChicagoIL60637USA
| | - Matthew V. Tirrell
- Pritzker School of Molecular EngineeringThe University of Chicago5640 S Ellis AveChicagoIL60637USA
| | - James L. LaBelle
- Department of Pediatrics, Section of Hematology/OncologyThe University of Chicago900 E 57th StChicagoIL60637USA
| |
Collapse
|
10
|
Zhang X, Wang DY, Wu X, Zhao Y, Li X, Ma R, Huang F, Shi L. “Spear and Shield in One” Nanochaperone Enables Protein to Navigate Multiple Biological Barriers for Enhanced Tumor Synergistic Therapy. Biomater Sci 2022; 10:3575-3584. [DOI: 10.1039/d2bm00409g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Protein therapeutics have been viewed as powerful candidates for cancer treatment by virtue of highly specific bioactivity and minimized adverse effects. However, the intracellular delivery of protein drugs remains enormously...
Collapse
|
11
|
Yang L, Fang H, Jiang J, Sha Y, Zhong Z, Meng F. EGFR-targeted pemetrexed therapy of malignant pleural mesothelioma. Drug Deliv Transl Res 2021; 12:2527-2536. [PMID: 34802094 DOI: 10.1007/s13346-021-01094-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2021] [Indexed: 12/17/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rare malignancy with poor prognosis, for which chemotherapy with pemetrexed (PEM) is among the few clinical treatments. PEM suffers, however, fast clearance, moderate drug exposure, and dose-limiting toxicities. Here, we report on epidermal growth factor receptor (EGFR)-targeted disulfide-crosslinked biodegradable chimaeric polymersomes (EGFR-CPs) to firmly load PEM and boost chemotherapy of MPM. EGFR-CPs encapsulating 8.7-16.4 wt.% PEM (EGFR-CPs-PEM) showed diameters of 62-65 nm and reduction-responsive drug release property. EGFR-CPs-PEM was more efficiently taken up by EGFR-overexpressed MSTO-211H cells, inducing about 4.7-fold enhanced anticancer activity compared with non-targeted CPs-PEM control. Intriguingly, the in vivo experiments in MSTO-211H xenograft mouse model revealed that EGFR-CPs-PEM brought about superior tumor deposition and penetration to CPs-PEM, and significantly more potent tumor repression than CPs-PEM and free PEM. This polymersome-enabled EGFR-targeted delivery of PEM offers an appealing therapeutic strategy for MPM.
Collapse
Affiliation(s)
- Liang Yang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Hanghang Fang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Jingjing Jiang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Yongjie Sha
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China.
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China.
| |
Collapse
|
12
|
Li Y, Zhou Y, Wang T, Long K, Zhang Y, Wang W. Photoenhanced cytosolic protein delivery based on a photocleavable group-modified dendrimer. NANOSCALE 2021; 13:17784-17792. [PMID: 34668505 DOI: 10.1039/d1nr04430c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Numerous recently developed therapies have highlighted the advantages of using proteins as therapeutics. However, in many protein delivery systems, the complicated carrier designs, low loading content, and off-targeting effects have limited their clinical applications. Here we report a photoresponsive protein-binding moiety and use it to prepare a simple nanoscale protein delivery system with high delivery efficiency and photoenhanced cellular uptake of proteins. The carrier was prepared by modifying a photocleavable molecule, DEACM, onto the surface of a cationic dendrimer, poly(amidoamine). DEACM simultaneously contributed to protein binding, self-assembly, and photocontrollability of the system. The multi-functional DEACM enabled the simplicity of the protein delivery system, which does not require complex organic synthesis or protein modification. The high delivery efficiency, high serum tolerance, and photoenhanced cellular uptake have been proved with functional proteins, presenting the potential for delivering protein therapeutics.
Collapse
Affiliation(s)
- Yafei Li
- State Key Laboratory of Pharmaceutical Biotechnology & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine & Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China.
| | - Yang Zhou
- State Key Laboratory of Pharmaceutical Biotechnology & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine & Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China.
| | - Tianyi Wang
- State Key Laboratory of Pharmaceutical Biotechnology & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine & Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China.
| | - Kaiqi Long
- State Key Laboratory of Pharmaceutical Biotechnology & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine & Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China.
| | - Yaming Zhang
- State Key Laboratory of Pharmaceutical Biotechnology & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine & Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China.
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical Biotechnology & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine & Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
13
|
Zhou H, Wang Y, Lu H. Intracellular delivery of His-tagged proteins via a hybrid organic–inorganic nanoparticle. Polym J 2021. [DOI: 10.1038/s41428-021-00526-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
14
|
Wei Y, Sun Y, Wei J, Qiu X, Meng F, Storm G, Zhong Z. Selective transferrin coating as a facile strategy to fabricate BBB-permeable and targeted vesicles for potent RNAi therapy of brain metastatic breast cancer in vivo. J Control Release 2021; 337:521-529. [PMID: 34352315 DOI: 10.1016/j.jconrel.2021.07.048] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/31/2021] [Accepted: 07/30/2021] [Indexed: 12/13/2022]
Abstract
Brain metastases are a most disturbing situation for breast cancer patients as there is basically no adequate treatment available. Any potential drug formulation has to be able to cross the blood-brain barrier (BBB) and specific to metastatic brain tumors without causing unacceptable adverse effects. Here, we developed transferrin-functionalized chimeric polymersomes carrying siRNA against polo-like kinase 1 (Tf@TBP-CPs-siPLK1) for treating brain metastatic MDA-MB 231 triple negative breast cancer (TNBC) xenografts in mice. To facilitate the loading of siPLK1, chimaeric polymersomes (CPs) were designed with spermine in the watery core and transferrin-binding peptide (TBP) at the surface, enabling attachment of transferrin after the siRNA loading step and thereby circumventing interference of transferrin with siRNA loading. Tf@TBP-CPs-siPLK1 encapsulating 3.8 wt% siRNA had a mean size of about 50 nm and a neutral zeta potential in phosphate buffer (PB). By virtue of the presence of transferrin, Tf@TBP-CPs demonstrated greatly (ca. 5-fold) enhanced internalization in MDA-MB 231 cells and transcytosis in the endothelial (bEnd.3) monolayer model in vitro as well as markedly improved accumulation in the orthotopically xenografted MDA-MB 231 tumor in the brain in vivo compared with control CPs lacking transferrin, supporting that transferrin mediates efficient BBB penetration and high specificity towards MDA-MB 231 cells. As a result, Tf@TBP-CPs-siPLK1 effectively inhibited tumor progression and prolonged the lifespan of the mice significantly. Selective transferrin coating appears to be a particularly facile strategy to fabricate BBB-permeable and targeted vesicles for potent RNAi therapy of brain metastatic breast cancer.
Collapse
Affiliation(s)
- Yaohua Wei
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Department of Biomaterials Science and Technology, MIRA Institute for Biological Technology and Technical Medicine, University of Twente, PO Box 217, 7500AE Enschede, the Netherlands
| | - Yinping Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Jingjing Wei
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Xinyun Qiu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| | - Gert Storm
- Department of Biomaterials Science and Technology, MIRA Institute for Biological Technology and Technical Medicine, University of Twente, PO Box 217, 7500AE Enschede, the Netherlands; Department of Pharmaceutics, Utrecht University, the Netherlands; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
15
|
Carrion CC, Nasrollahzadeh M, Sajjadi M, Jaleh B, Soufi GJ, Iravani S. Lignin, lipid, protein, hyaluronic acid, starch, cellulose, gum, pectin, alginate and chitosan-based nanomaterials for cancer nanotherapy: Challenges and opportunities. Int J Biol Macromol 2021; 178:193-228. [PMID: 33631269 DOI: 10.1016/j.ijbiomac.2021.02.123] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/07/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022]
Abstract
Although nanotechnology-driven drug delivery systems are relatively new, they are rapidly evolving since the nanomaterials are deployed as effective means of diagnosis and delivery of assorted therapeutic agents to targeted intracellular sites in a controlled release manner. Nanomedicine and nanoparticulate drug delivery systems are rapidly developing as they play crucial roles in the development of therapeutic strategies for various types of cancer and malignancy. Nevertheless, high costs, associated toxicity and production of complexities are some of the critical barriers for their applications. Green nanomedicines have continually been improved as one of the viable approaches towards tumor drug delivery, thus making a notable impact on which considerably affect cancer treatment. In this regard, the utilization of natural and renewable feedstocks as a starting point for the fabrication of nanosystems can considerably contribute to the development of green nanomedicines. Nanostructures and biopolymers derived from natural and biorenewable resources such as proteins, lipids, lignin, hyaluronic acid, starch, cellulose, gum, pectin, alginate, and chitosan play vital roles in the development of cancer nanotherapy, imaging and management. This review uncovers recent investigations on diverse nanoarchitectures fabricated from natural and renewable feedstocks for the controlled/sustained and targeted drug/gene delivery systems against cancers including an outlook on some of the scientific challenges and opportunities in this field. Various important natural biopolymers and nanomaterials for cancer nanotherapy are covered and the scientific challenges and opportunities in this field are reviewed.
Collapse
Affiliation(s)
- Carolina Carrillo Carrion
- Department of Organic Chemistry, University of Córdoba, Campus de Rabanales, Edificio Marie Curie, Ctra Nnal IV-A Km. 396, E-14014 Cordoba, Spain
| | | | - Mohaddeseh Sajjadi
- Department of Chemistry, Faculty of Science, University of Qom, Qom 37185-359, Iran
| | - Babak Jaleh
- Department of Physics, Bu-Ali Sina University, 65174 Hamedan, Iran
| | | | - Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
16
|
Wang H, Wang X, Zhang Y, Cheng R, Yuan J, Zhong Z. Systemic Delivery of NAC-1 siRNA by Neuropilin-Targeted Polymersomes Sensitizes Antiangiogenic Therapy of Metastatic Triple-Negative Breast Cancer. Biomacromolecules 2020; 21:5119-5127. [PMID: 33174734 DOI: 10.1021/acs.biomac.0c01253] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Antiangiogenic therapy with bevacizumab while being interesting for metastatic triple-negative breast cancer (mTNBC) is restrained by tumor hypoxia elevation and cancer stem cell enrichment. Here, we find that neuropilin-1 (NRP-1)-targeted delivery of nucleus accumbens-associated protein-1 (NAC-1) siRNA mediated by tLyP-1 peptide-functionalized chimaeric polymersomes (tLyP-1-Ps) effectively sensitizes antiangiogenic therapy of mTNBC in vivo. tLyP-1-Ps showed good encapsulation (up to 14.4 wt. %) of siNAC-1, giving robust tLyP-1-Ps-siNAC-1 nanoformulation with a defined size of 48.5 nm (PDI = 0.13) and a surface charge of -9.2 mV, and mediated efficient cytoplasmic transportation of siNAC-1 in MDA-MB-231 TNBC cells, resulting in significant silencing of NAC-1 mRNA and the corresponding oncoprotein. Transwell invasion and wound healing assays revealed that tLyP-1-Ps-siNAC-1 potently inhibited MDA-MB-231 cell invasion and migration. Intriguingly, tLyP-1-Ps-siNAC-1 was shown to markedly improve the bevacizumab therapy of mTNBC, significantly curbing lung metastasis and prolonging the survival time of the MDA-MB-231 metastatic model. The combination of targeted NAC-1 gene silencing and antiangiogenic therapy appears to be an innovative treatment for mTNBC.
Collapse
Affiliation(s)
- Hongyu Wang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Xiaohui Wang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Yi Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Ru Cheng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Jiandong Yuan
- BrightGene Bio-Medical Technology Company, Ltd., Suzhou 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
17
|
Zhao T, Fu Y, Jang MS, Sun XS, Wu T, Lee JH, Li Y, Lee DS, Yang HY. A pH-activated charge convertible quantum dot as a novel nanocarrier for targeted protein delivery and real-time cancer cell imaging. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111449. [PMID: 33255037 DOI: 10.1016/j.msec.2020.111449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/06/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
The rapid developments of nanocarriers based on quantum dots (QDs) have been confirmed to show substantial promise for drug delivery and bioimaging. However, optimal QDs-based nanocarriers still need to have their controlled behavior in vitro and in vivo and decrease heavy metal-associated cytotoxicity. Herein, a pH-activated charge convertible QD-based nanocarrier was fabricated by capping multifunctional polypeptide ligands (mPEG-block-poly(ethylenediamine-dihydrolipoic acid-2,3-dimethylmaleic anhydride)-L-glutamate, PEG-P(ED-DLA-DMA)LG) onto the surface of core/multishell CdSe@ZnS/ZnS QD by means of a ligand exchange strategy, followed by uploading of cytochrome C (CC) (CC-loaded QD-PEG-P(ED-DLA-DMA)LG) via electrostatic interactions, in which QDs that were water-soluble and protein-loading were perfectly integrated. That is, the CC-loaded QD-PEG-P(ED-DLA-DMA)LG inherited excellent fluorescence properties from CdSe@ZnS/ZnS QD for real-time imaging, as well as tumor-microenvironment sensitivities from PEG-P(ED-DLA-DMA)LG for enhanced cellular uptake and CC release. Experimental results verified that the QD-PEG-P(ED-DLA-DMA)LG showed enhanced internalization, rapid endo/lysosomal escape, and supplied legible real-time imaging for lung carcinoma cells. Furthermore, pH-triggered charge-convertible ability enabled the QD-PEG-P(ED-DLA-DMA)LG-CC to effectively kill cancer cells better than did the control groups. Hence, constructing smart nanocomposites by facile ligand-exchange strategy is beneficial to QD-based nanocarrier for tumor-targeting cancer therapy.
Collapse
Affiliation(s)
- Ting Zhao
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, People's Republic of China; College of Chemistry, Jilin University, Changchun City 130012, People's Republic of China
| | - Yan Fu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, People's Republic of China
| | - Moon-Sun Jang
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Xin Shun Sun
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, People's Republic of China
| | - Tepeng Wu
- Theranostic Macromolecules Research Center and School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Jung Hee Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Yi Li
- Theranostic Macromolecules Research Center and School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Doo Sung Lee
- Theranostic Macromolecules Research Center and School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Hong Yu Yang
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, People's Republic of China.
| |
Collapse
|
18
|
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
|
19
|
Abstract
Peptides are one of the most important functional motifs for constructing smart drug delivery systems (DDSs). Functional peptides can be conjugated with drugs or carriers via covalent bonds, or assembled into DDSs via supramolecular forces, which enables the DDSs to acquire desired functions such as targeting and/or environmental responsiveness. In this mini review, we first introduce the different types of functional peptides that are commonly used for constructing DDSs, and we highlight representative strategies for designing smart DDSs by using functional peptides in the past few years. We also state the challenges of peptide-based DDSs and come up with prospects.
Collapse
Affiliation(s)
- Zheng Lian
- People's Public Security University of China, Beijing 100038, China
| | | |
Collapse
|
20
|
Jäger E, Sincari V, Albuquerque LJC, Jäger A, Humajova J, Kucka J, Pankrac J, Paral P, Heizer T, Janouskova O, Konefał R, Pavlova E, Sedlacek O, Giacomelli FC, Pouckova P, Sefc L, Stepanek P, Hruby M. Reactive Oxygen Species (ROS)-Responsive Polymersomes with Site-Specific Chemotherapeutic Delivery into Tumors via Spacer Design Chemistry. Biomacromolecules 2020; 21:1437-1449. [PMID: 32083473 DOI: 10.1021/acs.biomac.9b01748] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The lack of cellular and tissue specificities in conventional chemotherapies along with the generation of a complex tumor microenvironment (TME) limits the dosage of active agents that reaches tumor sites, thereby resulting in ineffective responses and side effects. Therefore, the development of selective TME-responsive nanomedicines is of due relevance toward successful chemotherapies, albeit challenging. In this framework, we have synthesized novel, ready-to-use ROS-responsive amphiphilic block copolymers (BCs) with two different spacer chemistry designs to connect a hydrophobic boronic ester-based ROS sensor to the polymer backbone. Hydrodynamic flow focusing nanoprecipitation microfluidics (MF) was used in the preparation of well-defined ROS-responsive PSs; these were further characterized by a combination of techniques [1H NMR, dynamic light scattering (DLS), static light scattering (SLS), transmission electron microscopy (TEM), and cryogenic TEM (cryo-TEM)]. The reaction with hydrogen peroxide releases an amphiphilic phenol or a hydrophilic carboxylic acid, which affects polymersome (PS) stability and cargo release. Therefore, the importance of the spacer chemistry in BC deprotection and PS stability and cargo release is herein highlighted. We have also evaluated the impact of spacer chemistry on the PS-specific release of the chemotherapeutic drug doxorubicin (DOX) into tumors in vitro and in vivo. We demonstrate that by spacer chemistry design one can enhance the efficacy of DOX treatments (decrease in tumor growth and prolonged animal survival) in mice bearing EL4 T cell lymphoma. Side effects (weight loss and cardiotoxicity) were also reduced compared to free DOX administration, highlighting the potential of the well-defined ROS-responsive PSs as TME-selective nanomedicines. The PSs could also find applications in other environments with high ROS levels, such as chronic inflammations, aging, diabetes, cardiovascular diseases, and obesity.
Collapse
Affiliation(s)
- Eliézer Jäger
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| | - Vladimir Sincari
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| | - Lindomar J C Albuquerque
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Avenida dos Estados 5001, Santo André 09210-580, Brazil
| | - Alessandro Jäger
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| | - Jana Humajova
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University in Prague, Salmovska 1, 120 00 Prague, Czech Republic
| | - Jan Kucka
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| | - Jan Pankrac
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, Prague 2, 120 00 Prague, Czech Republic
| | - Petr Paral
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, Prague 2, 120 00 Prague, Czech Republic
| | - Tomas Heizer
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, Prague 2, 120 00 Prague, Czech Republic
| | - Olga Janouskova
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| | - Rafał Konefał
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| | - Ewa Pavlova
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| | - Ondrej Sedlacek
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| | - Fernando C Giacomelli
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Avenida dos Estados 5001, Santo André 09210-580, Brazil
| | - Pavla Pouckova
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University in Prague, Salmovska 1, 120 00 Prague, Czech Republic
| | - Ludek Sefc
- Center for Advanced Preclinical Imaging (CAPI), First Faculty of Medicine, Charles University, Salmovská 3, Prague 2, 120 00 Prague, Czech Republic
| | - Petr Stepanek
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| | - Martin Hruby
- Institute of Macromolecular Chemistry, Heyrovsky Sq. 2, 162 06 Prague, Czech Republic
| |
Collapse
|
21
|
Iqbal S, Blenner M, Alexander-Bryant A, Larsen J. Polymersomes for Therapeutic Delivery of Protein and Nucleic Acid Macromolecules: From Design to Therapeutic Applications. Biomacromolecules 2020; 21:1327-1350. [DOI: 10.1021/acs.biomac.9b01754] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Shoaib Iqbal
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
- Department of Bioengineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Mark Blenner
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Angela Alexander-Bryant
- Department of Bioengineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Jessica Larsen
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
- Department of Bioengineering, Clemson University, Clemson, South Carolina 29634, United States
| |
Collapse
|
22
|
Pottanam Chali S, Ravoo BJ. Polymer Nanocontainers for Intracellular Delivery. Angew Chem Int Ed Engl 2020; 59:2962-2972. [PMID: 31364243 PMCID: PMC7028112 DOI: 10.1002/anie.201907484] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/26/2019] [Indexed: 01/05/2023]
Abstract
Carriers for intracellular delivery are required to overcome limitations of therapeutic agents such as low specificity, systemic toxicity, high clearance rate, and low therapeutic index. Nanocontainers comprised of an aqueous core and a polymer shell have received increasing attention because they readily combine stimuli response to improve intracellular payload release and surface modification to enhance selectivity towards the desired region of action. This Minireview summarizes the design and properties of polymer nanocontainers for intracellular delivery, classified according to the polymer architecture.
Collapse
Affiliation(s)
- Sharafudheen Pottanam Chali
- Organic Chemistry Institute and Centre for Soft NanoscienceWestfälische Wilhelms-Universität MünsterCorrensstrasse 4048149MünsterGermany
| | - Bart Jan Ravoo
- Organic Chemistry Institute and Centre for Soft NanoscienceWestfälische Wilhelms-Universität MünsterCorrensstrasse 4048149MünsterGermany
| |
Collapse
|
23
|
Meyer CE, Abram SL, Craciun I, Palivan CG. Biomolecule–polymer hybrid compartments: combining the best of both worlds. Phys Chem Chem Phys 2020; 22:11197-11218. [DOI: 10.1039/d0cp00693a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent advances in bio/polymer hybrid compartments in the quest to obtain artificial cells, biosensors and catalytic compartments.
Collapse
Affiliation(s)
| | | | - Ioana Craciun
- Department of Chemistry
- University of Basel
- Basel
- Switzerland
| | | |
Collapse
|
24
|
Yang HY, Li Y, Jang MS, Fu Y, Wu T, Lee JH, Lee DS. Green preparation of pH-responsive and dual targeting hyaluronic acid nanogels for efficient protein delivery. Eur Polym J 2019. [DOI: 10.1016/j.eurpolymj.2019.109342] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
25
|
Qin X, Yu C, Wei J, Li L, Zhang C, Wu Q, Liu J, Yao SQ, Huang W. Rational Design of Nanocarriers for Intracellular Protein Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902791. [PMID: 31496027 DOI: 10.1002/adma.201902791] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/04/2019] [Indexed: 06/10/2023]
Abstract
Protein/antibody therapeutics have exhibited the advantages of high specificity and activity even at an extremely low concentration compared to small molecule drugs. However, they are accompanied by unfavorable physicochemical properties such as fragile tertiary structure, large molecular size, and poor penetration of the membrane, and thus the clinical use of protein drugs is hindered by inefficient delivery of proteins into the host cells. To overcome the challenges associated with protein therapeutics and enhance their biopharmaceutical applications, various protein-loaded nanocarriers with desired functions, such as lipid nanocapsules, polymeric nanoparticles, inorganic nanoparticles, and peptides, are developed. In this review, the different strategies for intracellular delivery of proteins are comprehensively summarized. Their designed routes, mechanisms of action, and potential therapeutics in live cells or in vivo are discussed in detail. Furthermore, the perspective on the new generation of delivery systems toward the emerging area of protein-based therapeutics is presented as well.
Collapse
Affiliation(s)
- Xiaofei Qin
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Changmin Yu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Jing Wei
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Chengwu Zhang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Qiong Wu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Jinhua Liu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211800, P. R. China
- Shaanxi Institute of Flexible Electronics (SIFE), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, P. R. China
| |
Collapse
|
26
|
Multifunctional hyaluronic acid-mediated quantum dots for targeted intracellular protein delivery and real-time fluorescence imaging. Carbohydr Polym 2019; 224:115174. [DOI: 10.1016/j.carbpol.2019.115174] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/17/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022]
|
27
|
Pottanam Chali S, Ravoo BJ. Polymernanocontainer für den Transport in das Zellinnere. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201907484] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sharafudheen Pottanam Chali
- Organisch-Chemisches Institut und Center for Soft Nanoscience Westfälische Wilhelms-Universität Münster Corrensstraße 40 48149 Münster Deutschland
| | - Bart Jan Ravoo
- Organisch-Chemisches Institut und Center for Soft Nanoscience Westfälische Wilhelms-Universität Münster Corrensstraße 40 48149 Münster Deutschland
| |
Collapse
|
28
|
Lv J, Fan Q, Wang H, Cheng Y. Polymers for cytosolic protein delivery. Biomaterials 2019; 218:119358. [DOI: 10.1016/j.biomaterials.2019.119358] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/11/2019] [Accepted: 07/13/2019] [Indexed: 12/31/2022]
|
29
|
Ding L, Gu W, Zhang Y, Yue S, Sun H, Cornelissen JJLM, Zhong Z. HER2-Specific Reduction-Sensitive Immunopolymersomes with High Loading of Epirubicin for Targeted Treatment of Ovarian Tumor. Biomacromolecules 2019; 20:3855-3863. [DOI: 10.1021/acs.biomac.9b00947] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Lin Ding
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Wenxing Gu
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
- Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE Enschede, The Netherlands
| | - Yifan Zhang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Shujing Yue
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Huanli Sun
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Jeroen J. L. M. Cornelissen
- Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE Enschede, The Netherlands
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
30
|
Qiu M, Glass Z, Xu Q. Nonviral Nanoparticles for CRISPR-Based Genome Editing: Is It Just a Simple Adaption of What Have Been Developed for Nucleic Acid Delivery? Biomacromolecules 2019; 20:3333-3339. [PMID: 31342740 DOI: 10.1021/acs.biomac.9b00783] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Genome-editing technologies hold tremendous potential for treating genetic diseases. However, the efficient and safe delivery of genome-editing elements to the location of interest, and the achievement of specific targeted gene correction without off-target side effect remains a big challenge. In this Perspective, we highlight recent developments and discuss the challenges of nonviral nanoparticles for the delivery of genome-editing tools. Finally, we will propose promising strategies to improve the delivery efficacy and advance the clinical translation of gene-editing technology.
Collapse
Affiliation(s)
- Min Qiu
- Department of Biomedical Engineering , Tufts University , 4 Colby Street , Medford , Massachusetts 02155 , United States
| | - Zachary Glass
- Department of Biomedical Engineering , Tufts University , 4 Colby Street , Medford , Massachusetts 02155 , United States
| | - Qiaobing Xu
- Department of Biomedical Engineering , Tufts University , 4 Colby Street , Medford , Massachusetts 02155 , United States
| |
Collapse
|
31
|
Huang K, He Y, Zhu Z, Guo J, Wang G, Deng C, Zhong Z. Small, Traceable, Endosome-Disrupting, and Bioresponsive Click Nanogels Fabricated via Microfluidics for CD44-Targeted Cytoplasmic Delivery of Therapeutic Proteins. ACS APPLIED MATERIALS & INTERFACES 2019; 11:22171-22180. [PMID: 31190543 DOI: 10.1021/acsami.9b05827] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Nanogels (NG) are among the most ideal cytoplasmic protein delivery vehicles; however, their performance is suboptimal, partly owing to relatively big size, poor cell uptake, and endosomal entrapment. Here, we developed small, traceable, endosome-disrupting, and bioresponsive hyaluronic acid NG (HA-NG) for CD44-targeted intracellular delivery of therapeutic proteins. With microfluidics and catalyst-free photo-click cross-linking, HA-NG with hydrodynamic diameters of ca. 80 and 150 nm, strong green fluorescence and efficient loading of various proteins including saporin (Sap), cytochrome C, herceptin, immunoglobulin G (IgG), and bovine serum albumin could be fabricated. Interestingly, 80 nm-sized HA-NG revealed clearly better cellular uptake than its 150 nm counterparts in both CD44-negative U87 cancer cells and CD44-positive 4T1 and MDA-MB-231 cells. Moreover, small NG exhibited accelerated endosomal escape, which was further boosted by introducing GALA, a pH-sensitive fusogenic peptide. Accordingly, Sap-loaded small and GALA-functionalized HA-NG showed the highest cytotoxicity in CD44-positive MDA-MB-231, 4T1, A549, and SMMC-7721 cancer cells. The biodistribution studies demonstrated that 80 nm-sized HA-NG displayed significantly greater tumor uptake as well as penetration in MDA-MB-231 human breast tumor xenografts than its 150 nm counterparts, whereas the introduction of GALA had no detrimental effect on tumor accumulation. Small, endosome-disrupting, and bioresponsive HA-NG with easy and controlled fabrication hold a great potential for targeted protein therapy.
Collapse
Affiliation(s)
- Ke Huang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| | - Yahui He
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| | - Zhehong Zhu
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| | - Jiakun Guo
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| | - Guanglin Wang
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences , Medical College of Soochow University , Suzhou 215123 , China
| | - Chao Deng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , China
| |
Collapse
|
32
|
Lecommandoux S, Klok HA, Zhong Z, Deming TJ. Future Directions at the Frontier of Polymer Science and Biology. Biomacromolecules 2019; 20:1-3. [PMID: 30990714 DOI: 10.1021/acs.biomac.8b01785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
| | - Harm-Anton Klok
- École Polytechnique Fédérale de Lausanne (EPFL), Institut des Matériaux and Institut des Sciences et Ingeńierie Chimiques , Laboratoire des Polymères, Bâtiment MXD, Station 12, CH-1015 Lausanne , Switzerland
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science , Soochow University , Suzhou 215123 , People's Republic of China
| | - Timothy J Deming
- Department of Chemistry and Biochemistry, and Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
| |
Collapse
|
33
|
Affiliation(s)
- Xun Liu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Fan Wu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Yong Ji
- Department of Cardiothoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| |
Collapse
|