1
|
Mirhadi E, Butler AE, Kesharwani P, Sahebkar A. Utilizing stimuli-responsive nanoparticles to deliver and enhance the anti-tumor effects of bilirubin. Biotechnol Adv 2024; 77:108469. [PMID: 39427964 DOI: 10.1016/j.biotechadv.2024.108469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/12/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Bilirubin (BR) is among the most potent endogenous antioxidants that originates from the heme catabolic pathway. Despite being considered as a dangerous and cytotoxic waste product at high concentrations, BR has potent antioxidant effects leading to the reduction of oxidative stress and inflammation, which play an important role in the development and progression of cancer. The purpose of this study is to introduce PEGylated BR nanoparticles (NPs), themselves or in combination with other anti-cancer agents. BR is effective when loaded into various nanoparticles and used in cancer therapy. Interestingly, BRNPs can be manipulated to create stimuli-responsive carriers providing a sustained and controlled, as well as on-demand, release of drug in response to internal or external factors such as reactive oxygen species, glutathione, light, enzymes, and acidic pH. This review suggests that BRNPs have the potential as tumor microenvironment-responsive delivery systems for effective targeting of various types of cancers.
Collapse
Affiliation(s)
- Elaheh Mirhadi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Wang Y, Zou R, Zhou Y, Zheng Y, Peng C, Liu Y, Tan H, Fu Q, Ding M. Unraveling mechanisms of protein encapsulation and release in coacervates via molecular dynamics and machine learning. Chem Sci 2024; 15:13442-13451. [PMID: 39183928 PMCID: PMC11339950 DOI: 10.1039/d4sc03061c] [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: 05/10/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Coacervates play a pivotal role in protein-based drug delivery research, yet their drug encapsulation and release mechanisms remain poorly understood. Here, we utilized the Martini model to investigate bovine serum albumin (BSA) protein encapsulation and release within polylysine/polyglutamate (PLys/PGlu) coacervates. Our findings emphasize the importance of ingredient addition sequence in coacervate formation and encapsulation rates, attributed to preference contact between oppositely charged proteins and poly(amino acid)s. Notably, coacervates composed of β-sheet poly(amino acid)s demonstrate greater BSA encapsulation efficiency due to their reduced entropy and flexibility. Furthermore, we examined the pH responsiveness of coacervates, shedding light on the dissolution process driven by Coulomb forces. By leveraging machine learning algorithms to analyze simulation results, our research advances the understanding of coacervate-based drug delivery systems, with the ultimate goal of optimizing therapeutic outcomes.
Collapse
Affiliation(s)
- Yiwei Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Rongrong Zou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Yeqiang Zhou
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Yi Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Chuan Peng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Yang Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Qiang Fu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| | - Mingming Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University Chengdu 610065 China
| |
Collapse
|
3
|
Fan Z, Iqbal H, Ni J, Khan NU, Irshad S, Razzaq A, Alfaifi MY, Elbehairi SEI, Shati AA, Zhou J, Cheng H. Rationalized landscape on protein-based cancer nanomedicine: Recent progress and challenges. Int J Pharm X 2024; 7:100238. [PMID: 38511068 PMCID: PMC10951516 DOI: 10.1016/j.ijpx.2024.100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
The clinical advancement of protein-based nanomedicine has revolutionized medical professionals' perspectives on cancer therapy. Protein-based nanoparticles have been exploited as attractive vehicles for cancer nanomedicine due to their unique properties derived from naturally biomacromolecules with superior biocompatibility and pharmaceutical features. Furthermore, the successful translation of Abraxane™ (paclitaxel-based albumin nanoparticles) into clinical application opened a new avenue for protein-based cancer nanomedicine. In this mini-review article, we demonstrate the rational design and recent progress of protein-based nanoparticles along with their applications in cancer diagnosis and therapy from recent literature. The current challenges and hurdles that hinder clinical application of protein-based nanoparticles are highlighted. Finally, future perspectives for translating protein-based nanoparticles into clinic are identified.
Collapse
Affiliation(s)
- Zhechen Fan
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Haroon Iqbal
- Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jiang Ni
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi 214000, China
| | - Naveed Ullah Khan
- Department of Pharmacy, Zhejiang University of Technology, Hangzhou 310000, China
| | - Shahla Irshad
- Department of Allied Health Sciences, Faculty of Health and Medical Sciences, Mirpur University of Science and Technology (MUST), Mirpur, Azad Jammu and Kashmir 10250, Pakistan
| | - Anam Razzaq
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mohammad Y. Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | | | - Ali A. Shati
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
4
|
Hua C, Qiu L. Polymersomes for Therapeutic Protein and Peptide Delivery: Towards Better Loading Properties. Int J Nanomedicine 2024; 19:2317-2340. [PMID: 38476284 PMCID: PMC10929215 DOI: 10.2147/ijn.s444910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/24/2024] [Indexed: 03/14/2024] Open
Abstract
Therapeutics based on proteins and peptides have profoundly transformed the landscape of treatment for diseases, from diabetes mellitus to cancers, yet the short half-life and low bioavailability of therapeutic proteins and peptides hinder their wide applications. To break through this bottleneck, biomolecules-loaded polymersomes with strong adjustability and versatility have attracted more and more attentions recently. Loading proteins or peptides into polymersomes is the first but extremely important step towards developing high-quality formulation products. However, increasing protein and peptide loading content is quite challenging due to the inherent nature of self-assembled vesicle formation mechanism and physiochemical characteristics of biomacromolecules. This review highlights the potential of polymersomes as the next-generation therapeutic proteins and peptides carrier and emphatically introduces novel approaches and recent progress to achieve satisfactory encapsulation capability of polymersomes for proteins and peptides. On the one hand, with the help of intermolecular interactions, such as electrostatic, lipid-protein, and hydrophobic interactions, the drug loading could be significantly improved. On the other hand, loading improvement could be attained through innovation of preparation methods, ranging from modified traditional film hydration techniques to the novel phase-guided assembly method.
Collapse
Affiliation(s)
- Chengxu Hua
- Ministry of Educational (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, People’s Republic of China
| | - Liyan Qiu
- Ministry of Educational (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, People’s Republic of China
| |
Collapse
|
5
|
Sinsinbar G, Bindra AK, Liu S, Chia TW, Yoong Eng EC, Loo SY, Lam JH, Schultheis K, Nallani M. Amphiphilic Block Copolymer Nanostructures as a Tunable Delivery Platform: Perspective and Framework for the Future Drug Product Development. Biomacromolecules 2024; 25:541-563. [PMID: 38240244 DOI: 10.1021/acs.biomac.3c00858] [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: 02/13/2024]
Abstract
Nanoformulation of active payloads or pharmaceutical ingredients (APIs) has always been an area of interest to achieve targeted, sustained, and efficacious delivery. Various delivery platforms have been explored, but loading and delivery of APIs have been challenging because of the chemical and structural properties of these molecules. Polymersomes made from amphiphilic block copolymers (ABCPs) have shown enormous promise as a tunable API delivery platform and confer multifold advantages over lipid-based systems. For example, a COVID booster vaccine comprising polymersomes encapsulating spike protein (ACM-001) has recently completed a Phase I clinical trial and provides a case for developing safe drug products based on ABCP delivery platforms. However, several limitations need to be resolved before they can reach their full potential. In this Perspective, we would like to highlight such aspects requiring further development for translating an ABCP-based delivery platform from a proof of concept to a viable commercial product.
Collapse
Affiliation(s)
- Gaurav Sinsinbar
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Anivind Kaur Bindra
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Shaoqiong Liu
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Teck Wan Chia
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Eunice Chia Yoong Eng
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Ser Yue Loo
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Jian Hang Lam
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Katherine Schultheis
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| | - Madhavan Nallani
- ACM Biolabs Pte Ltd., 71 Nanyang Drive, #02M-02, NTU Innovation Center, Singapore 638075, Singapore
| |
Collapse
|
6
|
Zhang F, Yao Q, Chen X, Zhou H, Zhou M, Li Y, Cheng H. In-depth study of anticancer drug diffusion through a cross-linked -pH-responsive polymeric vesicle membrane. Drug Deliv 2023; 30:2162626. [PMID: 36600638 PMCID: PMC9828689 DOI: 10.1080/10717544.2022.2162626] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Post-encapsulation and release of the anticancer drug doxorubicin hydrochloride (DOX·HCl) through cell-like transmission functions of polymeric vesicles were studied using cross-linked pH-responsive polymeric vesicles. The vesicles were fabricated for the first time via the redox-initiated reversible addition-fragmentation chain transfer dispersion polymerization in ethanol-water mixture, using 2-(diisopropylamino)ethyl methacrylate and glycidyl methacrylate, and the vesicle membrane was modified post-cross-linking by using ethylenediamine. A phase diagram was constructed for reproducible fabrication of the polymeric vesicles, and well-shaped vesicles were formed when the target degree of polymerization of the hydrophobic polymer chains was equal to or higher than 50 with solid content in the range of 10-30 wt%. The cross-linked vesicle membrane served as a gate enabling "open" and "closed" states in response to pH stimulation. Up to 50% drug loading efficiency and 39% drug loading content could be achieved, and in vitro release of the DOX-loaded vesicles in aqueous buffer solutions showed a much faster DOX release rate at pH 5.0 than at pH 6.5. The polymeric vesicles were of very low cytotoxicity to A549 cells up to the concentration of 2 mg/mL, and the IC50 of DOX-loaded vesicles were higher than that of the free DOX. The intracellular DOX release study indicated higher cellular uptake capability for DOX-loaded vesicles than that of free DOX.
Collapse
Affiliation(s)
- Fen Zhang
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China,CONTACT Fen Zhang ; Yantao Li Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province050081, China; Hua Cheng Institute of Biology, Hebei Academy of Sciences, Shijiazhuang, Hebei Province050081, China
| | - Qian Yao
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Xiaoqi Chen
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Haijun Zhou
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Mengmeng Zhou
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Yantao Li
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China,CONTACT Fen Zhang ; Yantao Li Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province050081, China; Hua Cheng Institute of Biology, Hebei Academy of Sciences, Shijiazhuang, Hebei Province050081, China
| | - Hua Cheng
- Institute of Biology, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China,CONTACT Fen Zhang ; Yantao Li Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province050081, China; Hua Cheng Institute of Biology, Hebei Academy of Sciences, Shijiazhuang, Hebei Province050081, China
| |
Collapse
|
7
|
Kayani A, Raza A, Si J, Dutta D, Zhou Q, Ge Z. Polymersome Membrane Engineering with Active Targeting or Controlled Permeability for Responsive Drug Delivery. Biomacromolecules 2023; 24:4622-4645. [PMID: 37870458 DOI: 10.1021/acs.biomac.3c00839] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Polymersomes have been extensively investigated for drug delivery as nanocarriers for two decades due to a series of advantages including high stability under physiological conditions, simultaneous encapsulation of hydrophilic and hydrophobic drugs inside inner cavities and membranes, respectively, and facile adjustment of membrane and surface properties, as well as controlled drug release through incorporation of stimuli-responsive components. Despite these features, polymersome nanocarriers frequently suffer from nontargeting delivery and poor membrane permeability. In recent years, polymersomes have been functionalized for more efficient drug delivery. The surface shells were explored to be modified with diverse active targeting groups to improve disease-targeting delivery. The membrane permeability of the polymersomes was adjusted by incorporation of the stimuli-responsive components for smart controlled transportation of the encapsulated drugs. Therefore, being the polymersome-biointerface, tailorable properties can be introduced by its carefully modulated engineering. This review elaborates on the role of polymersome membranes as a platform to incorporate versatile features. First, we discuss how surface functionalization facilitates the directional journey to the targeting sites toward specific diseases, cells, or intracellular organelles via active targeting. Moreover, recent advances in the past decade related to membrane permeability to control drug release are also summarized. We finally discuss future development to promote polymersomes as in vivo drug delivery nanocarriers.
Collapse
Affiliation(s)
- Anum Kayani
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Arsalan Raza
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Jiale Si
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Debabrata Dutta
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Qinghao Zhou
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Zhishen Ge
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| |
Collapse
|
8
|
Wang D, Moreno S, Boye S, Voit B, Appelhans D. Crosslinked and Multi-Responsive Polymeric Vesicles as a Platform to Study Enzyme-Mediated Undocking Behavior: Toward Future Artificial Organelle Communication. Macromol Rapid Commun 2023; 44:e2200885. [PMID: 36755359 DOI: 10.1002/marc.202200885] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/19/2023] [Indexed: 02/10/2023]
Abstract
Various cellular functions are successfully mimicked, opening the door to the next generation of therapeutic approaches and systems biology. Herein, the first steps are taken toward the construction of artificial organelles for mimicking cell communication by docking and undocking of cargo in the membrane of swollen artificial organelles. Stimuli-responsive and crosslinked polymeric vesicles are used to allow docking processes at acidic pH at which ferrocene units in the swollen membrane state can undergo desired specific host-guest interaction using β-cyclodextrin as model cargo. The release of the cargo mediated by two different enzymes, glucose oxidase and α-amylase, is investigated, triggered by distinct enzymatic undocking mechanisms. Different release times for a useful transport are shown that can be adapted to different communication pathways. In addition, Förster resonance energy transfer (FRET) experiments further support the hypotheses of host-guest inclusion complexation formation and their time-dependent breakdown. This work paves a way to a platform based on polymeric vesicles for synthetic biology, cell functions mimicking, and the construction of multifunctional cargo delivery system.
Collapse
Affiliation(s)
- Dishi Wang
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
- Organic Chemistry of Polymers, Technische Universität Dresden, 01062, Dresden, Germany
| | - Silvia Moreno
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
| | - Susanne Boye
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
| | - Brigitte Voit
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
- Organic Chemistry of Polymers, Technische Universität Dresden, 01062, Dresden, Germany
| | - Dietmar Appelhans
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
| |
Collapse
|
9
|
Kotha R, Kara DD, Roychowdhury R, Tanvi K, Rathnanand M. Polymersomes Based Versatile Nanoplatforms for Controlled Drug Delivery and Imaging. Adv Pharm Bull 2023; 13:218-232. [PMID: 37342386 PMCID: PMC10278216 DOI: 10.34172/apb.2023.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/11/2021] [Accepted: 01/07/2022] [Indexed: 10/29/2023] Open
Abstract
Drug delivery systems made based on nanotechnology represent a novel drug carrier system that can change the face of therapeutics and diagnosis. Among all the available nanoforms polymersomes have wider applications due to their unique characteristic features like drug loading carriers for both hydrophilic and hydrophobic drugs, excellent biocompatibility, biodegradability, longer shelf life in the bloodstream and ease of surface modification by ligands. Polymersomes are defined as the artificial vesicles which are enclosed in a central aqueous cavity which are composed of self-assembly with a block of amphiphilic copolymer. Various techniques like film rehydration, direct hydration, nanoprecipitation, double emulsion technique and microfluidic technique are mostly used in formulating polymersomes employing different polymers like PEO-b-PLA, poly (fumaric/sebacic acid), poly(N-isopropylacrylamide) (PNIPAM), poly (dimethylsiloxane) (PDMS), and poly(butadiene) (PBD), PTMC-b-PGA (poly (dimethyl aminoethyl methacrylate)-b-poly(l-glutamic acid)) etc. Polymersomes have been extensively considered for the conveyance of therapeutic agents for diagnosis, targeting, treatment of cancer, diabetes etc. This review focuses on a comprehensive description of polymersomes with suitable case studies under the following headings: chemical structure, polymers used in the formulation, formulation methods, characterization methods and their application in the therapeutic, and medicinal filed.
Collapse
Affiliation(s)
- Rohini Kotha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal-576104, India
| | - Divya Dhatri Kara
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal-576104, India
| | - Rajeshwari Roychowdhury
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal-576104, India
| | - Katikala Tanvi
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal-576104, India
| | - Mahalaxmi Rathnanand
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal-576104, India
| |
Collapse
|
10
|
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: 31] [Impact Index Per Article: 31.0] [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
|
11
|
Cong X, Chen J, Xu R. Recent Progress in Bio-Responsive Drug Delivery Systems for Tumor Therapy. Front Bioeng Biotechnol 2022; 10:916952. [PMID: 35845404 PMCID: PMC9277442 DOI: 10.3389/fbioe.2022.916952] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/09/2022] [Indexed: 12/24/2022] Open
Abstract
Spatially- and/or temporally-controlled drug release has always been the pursuit of drug delivery systems (DDSs) to achieve the ideal therapeutic effect. The abnormal pathophysiological characteristics of the tumor microenvironment, including acidosis, overexpression of special enzymes, hypoxia, and high levels of ROS, GSH, and ATP, offer the possibility for the design of stimulus-responsive DDSs for controlled drug release to realize more efficient drug delivery and anti-tumor activity. With the help of these stimulus signals, responsive DDSs can realize controlled drug release more precisely within the local tumor site and decrease the injected dose and systemic toxicity. This review first describes the major pathophysiological characteristics of the tumor microenvironment, and highlights the recent cutting-edge advances in DDSs responding to the tumor pathophysiological environment for cancer therapy. Finally, the challenges and future directions of bio-responsive DDSs are discussed.
Collapse
Affiliation(s)
- Xiufeng Cong
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ran Xu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Ran Xu,
| |
Collapse
|
12
|
Wang T, Qin J, Cheng J, Li C, Du J. Intelligent design of polymersomes for antibacterial and anticancer applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1822. [PMID: 35673991 DOI: 10.1002/wnan.1822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 01/25/2023]
Abstract
Polymersomes (or polymer vesicles) have attracted much attention for biomedical applications in recent years because their lumen can be used for drug delivery and their coronas and membrane can be modified with a variety of functional groups. Thus, polymersomes are very suitable for improved antibacterial and anticancer therapy. This review mainly highlighted recent advances in the synthetic protocols and design principles of intelligent antibacterial and anticancer polymersomes. Antibacterial polymersomes are divided into three categories: polymersomes as antibiotic nanocarriers, intrinsically antibacterial polymersomes, and antibacterial polymersomes with supplementary means including photothermal and photodynamic therapy. Similarly, the anticancer polymersomes are divided into two categories: polymersomes-based delivery systems and anticancer polymersomes with supplementary means. In addition, the bilateral relationship between bacteria and cancer is addressed, since more and more evidences show that bacteria may cause cancer or promote cancer progression. Finally, prospective on next-generation antibacterial and anticancer polymersomes are discussed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Tao Wang
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China
| | - Jinlong Qin
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China.,Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiajing Cheng
- Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Li
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China.,Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
13
|
Chen S, Wu Y, Lortie F, Bernard J, Binder WH, Zhu J. Hydrogen-Bonds Mediated Nanomedicine: Design, Synthesis and Applications. Macromol Rapid Commun 2022; 43:e2200168. [PMID: 35609317 DOI: 10.1002/marc.202200168] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/30/2022] [Indexed: 11/08/2022]
Abstract
Among the various challenges in medicine, diagnosis, complete cure and healing of cancers remain difficult given the heterogeneity and complexity of such disease. Differing from conventional platforms with often unsatisfactory theranostic capabilities, the contribution of supramolecular interactions, such as hydrogen-bonds (H-bonds), to cancer nanotheranostics opens new perspectives for the design of biomedical materials, exhibiting remarkable properties and easier processability. Thanks to their dynamic characteristics, a feature generally observed for non-covalent interactions, H-bonding (macro)molecules can be used as supramolecular motifs for yielding drug- and diagnostic carriers that possess attractive features, arising from the combination of assembled nanoplatforms and the responsiveness of H-bonds. Thus H-bonded nanomedicine provides a rich toolbox that is useful to fulfill biomedical needs with unique advantages in early-stage diagnosis and therapy, demonstrating the promising potential in clinical translations and applications. We here summarize the design and synthetic routes towards H-bonded nanomedicines, focus on the growing understanding of the structure-function relationship for efficient cancer treatment. We propose a guidance for designing new H-bonded intelligent theranostic agents, to inspire more successful explorations of cancer nanotheranostics and finally to promote potential clinical translations. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Senbin Chen
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Yanggui Wu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| | - Frédéric Lortie
- Univ Lyon, CNRS, UMR 5223, Ingénierie des Matériaux Polymères, Université Claude Bernard Lyon1, INSA Lyon, Université Jean Monnet, Villeurbanne Cedex, F-69621, France
| | - Julien Bernard
- Univ Lyon, CNRS, UMR 5223, Ingénierie des Matériaux Polymères, Université Claude Bernard Lyon1, INSA Lyon, Université Jean Monnet, Villeurbanne Cedex, F-69621, France
| | - Wolfgang H Binder
- Chair of Macromolecular Chemistry, Faculty of Natural Science II (Chemistry, Physics and Mathematics), Martin Luther University Halle-Wittenberg, von-Danckelmann-Platz 4, Halle (Saale), D-06120, Germany
| | - Jintao Zhu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan, 430074, China
| |
Collapse
|
14
|
Tong PH, Zhu L, Zang Y, Li J, He XP, James TD. Metal-organic frameworks (MOFs) as host materials for the enhanced delivery of biomacromolecular therapeutics. Chem Commun (Camb) 2021; 57:12098-12110. [PMID: 34714306 DOI: 10.1039/d1cc05157a] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Biomacromolecular drugs have become an important class of therapeutic agents for the treatment of human diseases. Considering their high propensity for being degraded in the human body, the choice of an appropriate delivery system is key to ensure the therapeutic efficacy of biomacromolecular drugs in vivo. As an emerging class of supramolecular "host" materials, metal-organic frameworks (MOFs) exhibit advantages in terms of the tunability of pore size, encapsulation efficiency, controllable drug release, simplicity in surface functionalization and good biocompatibility. As a result, MOF-based host-guest systems have been extensively developed as a new class of flexible and powerful platform for the delivery of therapeutic biomacromolecules. In this review, we summarize current research progress in the synthesis of MOFs as delivery materials for a variety of biomacromolecules. Firstly, we briefly introduce the advances made in the use of biomacromolecular drugs for disease therapy and the types of commonly used clinical delivery systems. We then describe the advantages of using MOFs as delivery materials. Secondly, the strategies for the construction of MOF-encapsulated biomacromolecules (Biomacromolecules@MOFs) and the release mechanisms of the therapeutics are categorized. Thirdly, the application of MOFs to deliver different types of biomacromolecules (e.g., antigens/antibodies, enzymes, therapeutic proteins, DNA/RNA, polypeptides, and polysaccharides) for the treatment of various human diseases based on immunotherapy, gene therapy, starvation therapy and oxidation therapy is summarized. Finally, the remaining challenges and available opportunities for MOFs as drug delivery systems are outlined, which we anticipate will encourage additional research efforts directed towards developing Biomacromolecules@MOFs systems for biomedical applications.
Collapse
Affiliation(s)
- Pei-Hong Tong
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China.
| | - Ling Zhu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China.
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China.
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China.
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China.
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK. .,School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, 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: 14.7] [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
|
Shen J, Sun C, Wang Z, Chu Z, Liu C, Xu X, Xia M, Zhao M, Wang C. Sequential receptor-mediated mixed-charge nanomedicine to target pancreatic cancer, inducing immunogenic cell death and reshaping the tumor microenvironment. Int J Pharm 2021; 601:120553. [PMID: 33794325 DOI: 10.1016/j.ijpharm.2021.120553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/09/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer (PC) is an aggressive form of cancer with dense stroma and immune-suppressive microenvironment, which are the major barriers for treatment. To address such barriers, this study aimed to develop a sequential receptor-mediated mixed-charge targeted delivery system for PC based on 2-(3-((S)-5-amino-1-carboxypentyl)-ureido) pentanedioate (ACUPA-) and triphenylphosphonium (TPP+) modified nanomicelles containing ingenol-3-mebutate (I3A), which was named ACUPA-/TPP+-I3A or ACUPA/TPP-I3A. ACUPA/TPP-I3A induced immunogenic cell death (ICD), which significantly increased the number of tumor-infiltrating T lymphocytes, activated adaptive immunity, and achieved superior survival time. I3A, a novel anticancer drug, could induce PC cell necrosis to release damage-associated molecular patterns, thereby activating adaptive immunity. With certain ratios of negatively (ACUPA-) and positively (TPP+) charged ligands, ACUPA/TPP-I3A acquired a negative charge in plasma (pH 7.4, to inhibit aggregation and uptake in the circulation) and was neutral in the acidic tumor microenvironment (pH 5.0-6.0, to overcome electrostatic hindrances and facilitate transcytosis). Furthermore, neovascular endothelium-specific ACUPA enabled rapid transcytosis of ACUPA/TPP-I3A across tumor vessel walls, entering into endosome/lysosomes (pH 4.5-5.0, its charge became positive and exhibited lysosome escape). Then, ACUPA/TPP-I3A selectively targeted mitochondria, which correlated with TPP-mediated effect. Finally, I3A was released to induce ICD that activated adaptive immunity and achieved superior survival time. Therefore, reshaping of the tumor microenvironment and potentiating antitumor immunity using ACUPA-/TPP+-I3A constituted a novel strategy to prolong the survival time.
Collapse
Affiliation(s)
- Jingmin Shen
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Chao Sun
- Central Research Laboratory, Institute of Medical Science, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China
| | - Zhicheng Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Zhijie Chu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Chengcheng Liu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, 250021 Jinan, Shandong Province, China
| | - Xuelian Xu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Ming Xia
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, 250021 Jinan, Shandong Province, China
| | - Miaoqing Zhao
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, 250021 Jinan, Shandong Province, China.
| | - Cheng Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
17
|
Cohen L, Livney YD, Assaraf YG. Targeted nanomedicine modalities for prostate cancer treatment. Drug Resist Updat 2021; 56:100762. [PMID: 33857756 DOI: 10.1016/j.drup.2021.100762] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Abstract
Prostate cancer (PC) is the second most common cause of death amongst men in the USA. Therapy of PC has been transformed in the past decade by introducing novel therapeutics, advanced functional imaging and diagnostic approaches, next generation sequencing, as well as improved application of existing therapies in localized PC. Treatment of PC at the different stages of the disease may include surgery, androgen deprivation therapy (ADT), chemotherapy and radiation therapy. However, although ADT has proven efficacious in PC treatment, its effectiveness may be temporary, as these tumors frequently develop molecular mechanisms of therapy resistance, which allow them to survive and proliferate even under conditions of testosterone deprivation, inhibition of androgen receptor signaling, or cytotoxic drug treatment. Importantly, ADT was found to induce key alterations which frequently result in the formation of metastatic tumors displaying a therapy refractory phenotype. Hence, to overcome these serious therapeutic impediments, novel PC cell-targeted therapeutic strategies are being developed. These include diverse platforms enabling specific enhanced antitumor drug uptake and increased intracellular accumulation. Studies have shown that these novel treatment modalities lead to enhanced antitumor activity and diminished systemic toxicity due to the use of selective targeting and decreased drug doses. The underlying mechanism of targeting and internalization is based upon the interaction between a selective ligand, conjugated to a drug-loaded nanoparticle or directly to an anti-cancer drug, and a specific plasma membrane biomarker, uniquely overexpressed on the surface of PC cells. Another targeted therapeutic approach is the delivery of unique anti-oncogenic signaling pathway-based therapeutic drugs, which are selectively cytotoxic to PC cells. The current paper reviews PC targeted modalities reported in the past 6 years, and discusses both the advantages and limitations of the various targeted treatment strategies.
Collapse
Affiliation(s)
- Lital Cohen
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Yoav D Livney
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
18
|
Yu W, Maynard E, Chiaradia V, Arno MC, Dove AP. Aliphatic Polycarbonates from Cyclic Carbonate Monomers and Their Application as Biomaterials. Chem Rev 2021; 121:10865-10907. [DOI: 10.1021/acs.chemrev.0c00883] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Wei Yu
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Edward Maynard
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Viviane Chiaradia
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Maria C. Arno
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Andrew P. Dove
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| |
Collapse
|
19
|
Oz UC, Bolat ZB, Ozkose UU, Gulyuz S, Kucukturkmen B, Khalily MP, Ozcubukcu S, Yilmaz O, Telci D, Esendagli G, Sahin F, Bozkir A. A robust optimization approach for the breast cancer targeted design of PEtOx-b-PLA polymersomes. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:111929. [PMID: 33812571 DOI: 10.1016/j.msec.2021.111929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/19/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
The equipping of nanoparticles with the peptide moiety recognizing a particular receptor, enables cell or tissue-specific targeting, therefore the optimization of the targeted nanoparticles is a key factor in the formulation design process. In this paper, we report the optimization concept of Doxorubicin encapsulating PEtOx-b-PLA polymersome formulation equipped with Peptide18, which is a breast cancer recognizing tumor homing peptide, and the unveiling of the cell-specific delivery potential. The most dominant formulation parameters, which are the polymer to Doxorubicin mass ratio (w/w) and the aqueous to organic phase ratio (v/v), were optimized using Central Composite Design (CCD) based Response Surface Methodology. The characteristics of optimum polymersome formulation were determined as the hydrodynamic diameter of 146.35 nm, the PDI value of 0.136, and the encapsulation efficiency of 57.11% and TEM imaging, which are in agreement with the DLS data, showed the spherical morphology of the polymersomes. In order to demonstrate the breast cancer-specific delivery of targeted polymersomes, the flow cytometry and confocal microscopy analyses were carried out. The targeted polymersomes were accumulated 8 times higher in AU565 cells compared to MCF10A cells and the intracellular Doxorubicin was almost 10 times higher in AU565 cells. The CCD-mediated optimized targeted polymersomes proposed in this report holds the promise of targeted therapy for breast cancer and can be potentially used for the development of novel treatments.
Collapse
Affiliation(s)
- Umut Can Oz
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Yenimahalle, 06560, Ankara, Turkey
| | - Zeynep Busra Bolat
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, 26 Agustos Campus, 34755 Istanbul, Turkey; Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Kucukcekmece, 34303, Istanbul, Turkey
| | - Umut Ugur Ozkose
- Materials Institute, Marmara Research Center, TUBITAK, Gebze 41470, Kocaeli, Turkey; Department of Chemistry, Faculty of Science and Letters, Istanbul Technical University, Maslak 34469, Istanbul, Turkey; Department of Chemistry, Faculty of Science and Letters, Piri Reis University, Tuzla, 34940, Istanbul, Turkey
| | - Sevgi Gulyuz
- Materials Institute, Marmara Research Center, TUBITAK, Gebze 41470, Kocaeli, Turkey; Department of Chemistry, Faculty of Science and Letters, Istanbul Technical University, Maslak 34469, Istanbul, Turkey
| | - Berrin Kucukturkmen
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Yenimahalle, 06560, Ankara, Turkey
| | - Melek Parlak Khalily
- Department of Chemistry, Faculty of Science and Letters, Yozgat Bozok University, Yozgat 66200, Turkey
| | - Salih Ozcubukcu
- Department of Chemistry, Faculty of Science, Middle East Technical University, Ankara 06800, Turkey
| | - Ozgur Yilmaz
- Materials Institute, Marmara Research Center, TUBITAK, Gebze 41470, Kocaeli, Turkey
| | - Dilek Telci
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, 26 Agustos Campus, 34755 Istanbul, Turkey
| | - Gunes Esendagli
- Department of Basic Oncology, Cancer Institute, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, 26 Agustos Campus, 34755 Istanbul, Turkey
| | - Asuman Bozkir
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Yenimahalle, 06560, Ankara, Turkey.
| |
Collapse
|
20
|
Araste F, Aliabadi A, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. Self-assembled polymeric vesicles: Focus on polymersomes in cancer treatment. J Control Release 2021; 330:502-528. [DOI: 10.1016/j.jconrel.2020.12.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/16/2022]
|
21
|
Ni J, Miao T, Su M, Khan NU, Ju X, Chen H, Liu F, Han L. PSMA-targeted nanoparticles for specific penetration of blood-brain tumor barrier and combined therapy of brain metastases. J Control Release 2021; 329:934-947. [DOI: 10.1016/j.jconrel.2020.10.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
|
22
|
Gu W, Meng F, Haag R, Zhong Z. Actively targeted nanomedicines for precision cancer therapy: Concept, construction, challenges and clinical translation. J Control Release 2021; 329:676-695. [DOI: 10.1016/j.jconrel.2020.10.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/13/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023]
|
23
|
Gannimani R, Walvekar P, Naidu VR, Aminabhavi TM, Govender T. Acetal containing polymers as pH-responsive nano-drug delivery systems. J Control Release 2020; 328:736-761. [DOI: 10.1016/j.jconrel.2020.09.044] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/04/2023]
|
24
|
Torres J, Dhas N, Longhi M, García MC. Overcoming Biological Barriers With Block Copolymers-Based Self-Assembled Nanocarriers. Recent Advances in Delivery of Anticancer Therapeutics. Front Pharmacol 2020; 11:593197. [PMID: 33329001 PMCID: PMC7734332 DOI: 10.3389/fphar.2020.593197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/15/2020] [Indexed: 11/21/2022] Open
Abstract
Cancer is one of the most common life-threatening illness and it is the world's second largest cause of death. Chemotherapeutic anticancer drugs have many disadvantages, which led to the need to develop novel strategies to overcome these shortcomings. Moreover, tumors are heterogenous in nature and there are various biological barriers that assist in treatment reisistance. In this sense, nanotechnology has provided new strategies for delivery of anticancer therapeutics. Recently, delivery platforms for overcoming biological barriers raised by tumor cells and tumor-bearing hosts have been reported. Among them, amphiphilic block copolymers (ABC)-based self-assembled nanocarriers have attracted researchers worldwide owing to their unique properties. In this work, we addressed different biological barriers for effective cancer treatment along with several strategies to overcome them by using ABC-based self-assembled nanostructures, with special emphasis in those that have the ability to act as responsive nanocarriers to internal or external environmental clues to trigger release of the payload. These nanocarriers have shown promising properties to revolutionize cancer treatment and diagnosis, but there are still challenges for their successful translation to clinical applications.
Collapse
Affiliation(s)
- Jazmin Torres
- Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Namdev Dhas
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Marcela Longhi
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Mónica C. García
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| |
Collapse
|
25
|
Reinhard S, Han H, Tuma J, Røise JJ, Li IC, Li J, Lee HY, Murthy N. A pH-sensitive eosin-block copolymer delivers proteins intracellularly. Chem Commun (Camb) 2020; 56:14207-14210. [PMID: 33111731 PMCID: PMC10754351 DOI: 10.1039/d0cc05165a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
There is great interest in developing strategies to deliver proteins into the cytoplasm of cells. We report here a PEG-poly-eosin block copolymer (PEG-pEosin) that can encapsulate proteins and release them in active form under mildly acidic conditions. A PEG-pEosin formulation composed of Cre and the endosomolytic protein LLO efficiently performed gene editing in cells and in the brains of mice after an intracranial injection.
Collapse
Affiliation(s)
- Sören Reinhard
- Department of Bioengineering, University of California, and the Innovative Genomics Institute, 2151 Berkeley Way, Berkeley CA, 94720, USA.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Gao Z, Zhang Z, Guo J, Hao J, Zhang P, Cui J. Polypeptide Nanoparticles with pH-Sheddable PEGylation for Improved Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:13656-13662. [PMID: 33147977 DOI: 10.1021/acs.langmuir.0c02532] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The variation of tumor microenvironments provides a tool for the construction of stimulus-responsive nanomedicines to enhance drug delivery efficacy. Herein, the assembly of drug-loaded polypeptide nanoparticles (NPs) with pH-sheddable modification of poly(ethylene glycol) (PEG) is prepared to enhance therapeutic efficiency. Poly(l-lysine) and poly(l-glutamic acid) were self-assembled to fabricate polypeptide NPs by electrostatic interactions, followed by PEGylation based on amidation reaction. The NP sizes can be controlled by tuning the molecular weight or the ratio of polypeptides. The PEG coating is cleavable at the tumor acid microenvironment to reverse the surface charge and reduce the NP size, which effectively enhances cell uptake. In addition, the presence of reducing reagent (e.g., glutathione) in cancer cells induces the drug (i.e., cisplatin) release from the polypeptide NPs and subsequently results in the cell toxicity. This reported method highlights the engineering of transformable polypeptide drug carriers, which provides a promising way for enhanced drug delivery efficacy.
Collapse
Affiliation(s)
- Zhiliang Gao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Zhonghe Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
- Department of Medical Imaging, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Jianman Guo
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jingcheng Hao
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Peiyu Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong 266237, China
| |
Collapse
|
27
|
Xiao W, Jakimowicz MD, Zampetakis I, Neely S, Scarpa F, Davis SA, Williams DS, Perriman AW. Biopolymeric Coacervate Microvectors for the Delivery of Functional Proteins to Cells. ACTA ACUST UNITED AC 2020; 4:e2000101. [PMID: 33166084 DOI: 10.1002/adbi.202000101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/29/2020] [Indexed: 11/07/2022]
Abstract
The extent to which biologic payloads can be effectively delivered to cells is a limiting factor in the development of new therapies. Limitations arise from the lack of pharmacokinetic stability of biologics in vivo. Encapsulating biologics in a protective delivery vector has the potential to improve delivery profile and enhance performance. Coacervate microdroplets are developed as cell-mimetic materials with established potential for the stabilization of biological molecules, such as proteins and nucleic acids. Here, the development of biodegradable coacervate microvectors (comprising synthetically modified amylose polymers) is presented, for the delivery of biologic payloads to cells. Amylose-based coacervate microdroplets are stable under physiological conditions (e.g., temperature and ionic strength), are noncytotoxic owing to their biopolymeric structure, spontaneously interacted with the cell membrane, and are able to deliver and release proteinaceous payloads beyond the plasma membrane. In particular, myoglobin, an oxygen storage and antioxidant protein, is successfully delivered into human mesenchymal stem cells (hMSCs) within 24 h. Furthermore, coacervate microvectors are implemented for the delivery of human bone morphogenetic protein 2 growth factor, inducing differentiation of hMSCs into osteoprogenitor cells. This study demonstrates the potential of coacervate microdroplets as delivery microvectors for biomedical research and the development of new therapies.
Collapse
Affiliation(s)
- Wenjin Xiao
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Monika D Jakimowicz
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol, BS8 1FD, UK
- HH Wills Physics Laboratory, University of Bristol, Bristol, BS8 1TL, UK
- Centre for Organized Matter Chemistry and Centre for Protolife Research School of Chemistry, University of Bristol, Bristol, BS8 1TS, UK
| | - Ioannis Zampetakis
- Bristol Composites Institute (ACCIS), Department of Aerospace Engineering, University of Bristol, Bristol, BS8 1TF, UK
| | - Sarah Neely
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Fabrizio Scarpa
- Bristol Composites Institute (ACCIS), Department of Aerospace Engineering, University of Bristol, Bristol, BS8 1TF, UK
| | - Sean A Davis
- Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol, BS8 1FD, UK
| | - David S Williams
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| |
Collapse
|
28
|
Intracellular delivery of cytochrome C using hypoxia-responsive polypeptide micelles for efficient cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 114:111069. [DOI: 10.1016/j.msec.2020.111069] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 01/13/2023]
|
29
|
Yari H, Gali H, Awasthi V. Nanoparticles for Targeting of Prostate Cancer. Curr Pharm Des 2020; 26:5393-5413. [PMID: 32693761 DOI: 10.2174/1381612826666200721001500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/27/2020] [Indexed: 11/22/2022]
Abstract
Prostate cancer (PCa) is the leading cause of death by cancer in men. Because of the drastic decline in the survival rate of PCa patients with advanced/metastatic disease, early diagnosis of disease and therapy without toxic side effects is crucial. Chemotherapy is widely used to control the progression of PCa at the later stages; however, it is associated with off-target toxicities and severe adverse effects due to the lack of specificity. Delivery of therapeutic or diagnostic agents by using targeted nanoparticles is a promising strategy to enhance accuracy and sensitivity of diagnosis of PCa and to increase efficacy and specificity of therapeutic agents. Numerous efforts have been made in past decades to create nanoparticles with different architectural bases for specific delivery payloads to prostate tumors. Major PCa associated cell membrane protein markers identified as targets for such purposes include folate receptor, sigma receptors, transferrin receptor, gastrin-releasing peptide receptor, urokinase plasminogen activator receptor, and prostate specific membrane antigen. Among these markers, prostate specific membrane antigen has emerged as an extremely specific and sensitive targetable marker for designing targeted nanoparticle-based delivery systems for PCa. In this article, we review contemporary advances in design, specificity, and efficacy of nanoparticles functionalized against PCa. Whenever feasible, both diagnostic as well as therapeutic applications are discussed.
Collapse
Affiliation(s)
- Hooman Yari
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Hariprasad Gali
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
30
|
Emerging era of “somes”: polymersomes as versatile drug delivery carrier for cancer diagnostics and therapy. Drug Deliv Transl Res 2020; 10:1171-1190. [PMID: 32504410 DOI: 10.1007/s13346-020-00789-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the past two decades, polymersomes have been widely investigated for the delivery of diagnostic and therapeutic agents in cancer therapy. Polymersomes are stable polymeric vesicles, which are prepared using amphiphilic block polymers of different molecular weights. The use of high molecular weight amphiphilic copolymers allows for possible manipulation of membrane characteristics, which in turn enhances the efficiency of drug delivery. Polymersomes are more stable in comparison with liposomes and show less toxicity in vivo. Furthermore, their ability to encapsulate both hydrophilic and hydrophobic drugs, significant biocompatibility, robustness, high colloidal stability, and simple methods for ligands conjugation make polymersomes a promising candidate for therapeutic drug delivery in cancer therapy. This review is focused on current development in the application of polymersomes for cancer therapy and diagnosis. Graphical abstract.
Collapse
|
31
|
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: 22.3] [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
|
32
|
Peng Y, Bariwal J, Kumar V, Tan C, Mahato RI. Organic Nanocarriers for Delivery and Targeting of Therapeutic Agents for Cancer Treatment. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900136] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Peng
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Jitender Bariwal
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Virender Kumar
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug DeliveryUniversity of Mississippi University MS 38677 USA
| | - Ram I. Mahato
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| |
Collapse
|
33
|
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: 141] [Impact Index Per Article: 28.2] [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
|
34
|
Afsharzadeh M, Hashemi M, Babaei M, Abnous K, Ramezani M. PEG‐PLA nanoparticles decorated with small‐molecule PSMA ligand for targeted delivery of galbanic acid and docetaxel to prostate cancer cells. J Cell Physiol 2019; 235:4618-4630. [DOI: 10.1002/jcp.29339] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/30/2019] [Indexed: 01/15/2023]
Affiliation(s)
- Maryam Afsharzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Hashemi
- Nanotechnology Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Babaei
- Pharmaceutical Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
| | - Khalil Abnous
- Department of Medicinal Chemistry, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
35
|
Qian X, Shi Z, Qi H, Zhao M, Huang K, Han D, Zhou J, Liu C, Liu Y, Lu Y, Yuan X, Zhao J, Kang C. A novel Granzyme B nanoparticle delivery system simulates immune cell functions for suppression of solid tumors. Am J Cancer Res 2019; 9:7616-7627. [PMID: 31695790 PMCID: PMC6831455 DOI: 10.7150/thno.35900] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/25/2019] [Indexed: 12/26/2022] Open
Abstract
Cell-based immunotherapy for the treatment of hematologic malignancies, such as leukemia and lymphoma, has seen much success and played an increasingly important role in clinical studies. Nevertheless, the efficacy of immunotherapy in solid tumors still needs improvements due to the immunosuppressive properties of tumor cells and the microenvironment. To overcome these limitations, we prepared a novel tumor-targeting delivery system based on the underlying mechanism of immune-targeted cell death that encapsulated granzyme B protein within a porous polymeric nanocapsule. Methods: A cell-penetrating peptide TAT was attached onto granzyme B (GrB) to enhance its transmembrane transport efficiency and potency to induce cell apoptosis. The endocytosis and internalization pathways of GrB-TAT (GrB-T) were analyzed in comparison with perforin by confocal microscopy and flow cytometry. Furthermore, the positively charged GrB-T was wrapped into nanoparticles by p-2-methacryloyloxy ethyl phosphorylcholine (PMPC)-modified HA (hyaluronic acid). The nanoparticles (called TCiGNPs) were characterized in terms of zeta potential and by transmission electron microscopy (TEM). The in vitro anti-tumor effects of GrB-T were examined by cell apoptosis assay and Western blotting analysis. The in vivo anti-tumor therapeutic efficacy of TCiGNPs was evaluated in a mouse tumor model. Results: The TAT peptide could play a role similar to perforin to mediate direct transmembrane transfer of GrB and improve GrB-induced cell apoptosis. The TCiGNPs were successfully synthesized and accumulated in the solid tumor through enhanced permeability and retention (EPR) effect. In the tumor microenvironment, TCiGNPs could be degraded by hyaluronidase and triggered the release of GrB-T. The TAT peptide enabled the translocation of GrB across the plasma membrane to induce tumor cell apoptosis in vivo. Conclusion: We successfully developed a granzyme B delivery system with a GrB-T core and a PMPC/HA shell that simulated CTL/NK cell-mediated cancer immunotherapy mechanism. The GrB delivery system holds great promise for cancer treatment analogous to the CTL/NK cell-induced immunotherapy.
Collapse
|
36
|
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: 26.0] [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]
|
37
|
Geraldes DC, Beraldo-de-Araújo VL, Pardo BOP, Pessoa Junior A, Stephano MA, de Oliveira-Nascimento L. Protein drug delivery: current dosage form profile and formulation strategies. J Drug Target 2019; 28:339-355. [DOI: 10.1080/1061186x.2019.1669043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Danilo Costa Geraldes
- Faculty of Pharmaceutical Sciences, State University of Campinas, Campinas, SP, Brazil
- Biochemistry and Tissue Biology Department, Biology Institute, State University of Campinas, Campinas, SP, Brazil
| | - Viviane Lucia Beraldo-de-Araújo
- Faculty of Pharmaceutical Sciences, State University of Campinas, Campinas, SP, Brazil
- Biochemistry and Tissue Biology Department, Biology Institute, State University of Campinas, Campinas, SP, Brazil
| | | | | | | | - Laura de Oliveira-Nascimento
- Faculty of Pharmaceutical Sciences, State University of Campinas, Campinas, SP, Brazil
- Biochemistry and Tissue Biology Department, Biology Institute, State University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
38
|
Bamburowicz-Klimkowska M, Poplawska M, Grudzinski IP. Nanocomposites as biomolecules delivery agents in nanomedicine. J Nanobiotechnology 2019; 17:48. [PMID: 30943985 PMCID: PMC6448271 DOI: 10.1186/s12951-019-0479-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/18/2019] [Indexed: 02/08/2023] Open
Abstract
Nanoparticles (NPs) are atomic clusters of crystalline or amorphous structure that possess unique physical and chemical properties associated with a size range of between 1 and 100 nm. Their nano-sized dimensions, which are in the same range as those of vital biomolecules, such as antibodies, membrane receptors, nucleic acids, and proteins, allow them to interact with different structures within living organisms. Because of these features, numerous nanoparticles are used in medicine as delivery agents for biomolecules. However, off-target drug delivery can cause serious side effects to normal tissues and organs. Considering this issue, it is essential to develop bioengineering strategies to significantly reduce systemic toxicity and improve therapeutic effect. In contrast to passive delivery, nanosystems enable to obtain enhanced therapeutic efficacy, decrease the possibility of drug resistance, and reduce side effects of "conventional" therapy in cancers. The present review provides an overview of the most recent (mostly last 3 years) achievements related to different biomolecules used to enable targeting capabilities of highly diverse nanoparticles. These include monoclonal antibodies, receptor-specific peptides or proteins, deoxyribonucleic acids, ribonucleic acids, [DNA/RNA] aptamers, and small molecules such as folates, and even vitamins or carbohydrates.
Collapse
Affiliation(s)
| | - Magdalena Poplawska
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3 Str, 00-664, Warsaw, Poland
| | - Ireneusz P Grudzinski
- Department of Applied Toxicology, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str, 02-097, Warsaw, Poland.
| |
Collapse
|
39
|
Jiang Y, Zhang J, Meng F, Zhong Z. Apolipoprotein E Peptide-Directed Chimeric Polymersomes Mediate an Ultrahigh-Efficiency Targeted Protein Therapy for Glioblastoma. ACS NANO 2018; 12:11070-11079. [PMID: 30395440 DOI: 10.1021/acsnano.8b05265] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The inability to cross the blood-brain barrier (BBB) prevents nearly all chemotherapeutics and biotherapeutics from the effective treatment of brain tumors, rendering few improvements in patient survival rates to date. Here, we report that apolipoprotein E peptide [ApoE, (LRKLRKRLL)2C] specifically binds to low-density lipoprotein receptor members (LDLRs) and mediates superb BBB crossing and highly efficient glioblastoma (GBM)-targeted protein therapy in vivo. The in vitro BBB model studies reveal that ApoE induces 2.2-fold better penetration of the immortalized mouse brain endothelial cell line (bEnd.3) monolayer for chimeric polymersomes (CP) compared to Angiopep-2, the best-known BBB-crossing peptide used in clinical trials for GBM therapy. ApoE-installed CP (ApoE-CP) carrying saporin (SAP) displays a highly specific and potent antitumor effect toward U-87 MG cells with a low half-maximum inhibitory concentration of 14.2 nM SAP. Notably, ApoE-CP shows efficient BBB crossing as well as accumulation and penetration in orthotopic U-87 MG glioblastoma. The systemic administration of SAP-loaded ApoE-CP causes complete growth inhibition of orthotopic U-87 MG GBM without eliciting any observable adverse effects, affording markedly improved survival benefits. ApoE peptide provides an ultrahigh-efficiency targeting strategy for GBM therapy.
Collapse
Affiliation(s)
- Yu Jiang
- 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 , PR China
| | - Jian 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 , PR China
| | - Fenghua Meng
- 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 , PR 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 , PR China
| |
Collapse
|
40
|
Disease-directed design of biodegradable polymers: Reactive oxygen species and pH-responsive micellar nanoparticles for anticancer drug delivery. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2666-2677. [DOI: 10.1016/j.nano.2018.06.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/15/2018] [Accepted: 06/29/2018] [Indexed: 11/16/2022]
|
41
|
Pudlarz A, Szemraj J. Nanoparticles as Carriers of Proteins, Peptides and Other Therapeutic Molecules. Open Life Sci 2018; 13:285-298. [PMID: 33817095 PMCID: PMC7874720 DOI: 10.1515/biol-2018-0035] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/08/2018] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles have many applications both in industry and medicine. Depending upon their physical and chemical properties, they can be used as carriers of therapeutic molecules or as therapeutics. Nanoparticles are made of synthetic or natural polymers, lipids or metals. Their use allows for faster transport to the place of action, thus prolonging its presence in the body and limiting side effects. In addition, the use of such a drug delivery system protects the drug from rapid disintegration and elimination from the body. In recent years, the use of proteins and peptides as therapeutic molecules has grown significantly. Unfortunately, proteins are subject to enzymatic digestion and can cause unwanted immune response beyond therapeutic action. The use of drug carriers can minimize undesirable side effects and reduce the dose of medication needed to achieve the therapeutic effect. The current study presents the use of several selected drug delivery systems for the delivery of proteins, peptides and other therapeutic molecules.
Collapse
Affiliation(s)
- Agnieszka Pudlarz
- Department of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
- E-mail:
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
42
|
Yang W, Wei Y, Yang L, Zhang J, Zhong Z, Storm G, Meng F. Granzyme B-loaded, cell-selective penetrating and reduction-responsive polymersomes effectively inhibit progression of orthotopic human lung tumor in vivo. J Control Release 2018; 290:141-149. [PMID: 30312720 DOI: 10.1016/j.jconrel.2018.10.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/30/2018] [Accepted: 10/08/2018] [Indexed: 12/11/2022]
Abstract
The clinical use of protein therapeutics with intracellular targets is hampered by its in vivo fragility and low cell permeability. Here, we report that cell-selective penetrating and reduction-responsive polymersomes (CPRPs) mediate high-efficiency targeted delivery of granzyme B (GrB) to orthotopic human lung tumor in vivo. Model protein studies using FITC-labeled cytochrome C (FITC-CC) revealed efficient and high protein loading up to 17.2 wt% for CPRPs. FITC-CC-loaded CPRPs exhibited a small size of 82-90 nm, reduction-responsive protein release, as well as greatly enhanced internalization and cytoplasmic protein release in A549 lung cancer cells compared with the non-targeted FITC-CC-loaded RPs control. GrB-loaded CPRPs showed a high potency toward A549 lung cancer cells with a half maximal inhibitory concentration (IC50) of 20.7 nM. Under the same condition, free GrB was essentially non-toxic. Importantly, installing cell-selective penetrating peptide did not alter the circulation time but did enhance tumor accumulation of RPs. Orthotopic A549-Luc lung tumor-bearing nude mice administered with GrB-loaded CPRPs at a dosage of 2.88 nmol GrB equiv./kg showed complete tumor growth inhibition with little body weight loss throughout the treatment period, resulting in significantly improved survival rate over the non-targeted and non-treated controls. These cell-selective penetrating and reduction-responsive polymersomes provide a targeted protein therapy for cancers.
Collapse
Affiliation(s)
- Weijing Yang
- 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, PR China
| | - Yaohua Wei
- 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, PR China; Department of Targeted Therapeutics, MIRA Institute for Biological Technology and Technical Medicine, University of Twente, PO Box 217, Enschede 7500AE, The Netherlands
| | - Liang Yang
- 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, PR China
| | - Jian Zhang
- 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, PR 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, Soochow University, Suzhou 215123, PR China.
| | - Gert Storm
- Department of Targeted Therapeutics, MIRA Institute for Biological Technology and Technical Medicine, University of Twente, PO Box 217, Enschede 7500AE, The Netherlands
| | - Fenghua Meng
- 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, PR China.
| |
Collapse
|
43
|
Cheng L, Yang L, Meng F, Zhong Z. Protein Nanotherapeutics as an Emerging Modality for Cancer Therapy. Adv Healthc Mater 2018; 7:e1800685. [PMID: 30240152 DOI: 10.1002/adhm.201800685] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/31/2018] [Indexed: 12/22/2022]
Abstract
Protein drugs are a unique and versatile class of biotherapeutics that have not only high biological activity but also superb specificity. This rapidly evolving biotechnology has rendered it possible to produce various proteins in a large scale and reproducible way. Many proteins have demonstrated striking anticancer activities and have emerged as advanced alternatives to cytotoxic chemotherapeutic agents for cancer therapy. The clinical translation of anticancer proteins with intracellular targets is, nevertheless, severely hindered by their fast degradation in vivo, poor cell penetration, and inefficient intracellular transportation. The past few years have witnessed tremendous effort and progress in developing polymeric protein delivery nanosystems, ranging from nanoparticles, nanocapsules, nanogels, micelles, to polymersomes, for the treatment of different tumors such as lung tumors, breast tumors, ovarian cancers, and glioblastoma. These proof-of-concept studies point out that protein nanotherapeutics, with rationally designed nanovehicles, are able to overcome the extracellular barriers, cell membrane barriers, and intracellular barriers, and systemically deliver proteins into targeted cancer cells, resulting in effective cancer protein therapy. Protein nanotherapeutics appear to be a novel modality for safe and efficient cancer treatment.
Collapse
Affiliation(s)
- Liang Cheng
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
- 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, P. R. China
| | - Liang Yang
- 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, P. R. China
| | - Fenghua Meng
- 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, P. R. 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, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
44
|
Jiang Y, Yang W, Zhang J, Meng F, Zhong Z. Protein Toxin Chaperoned by LRP-1-Targeted Virus-Mimicking Vesicles Induces High-Efficiency Glioblastoma Therapy In Vivo. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1800316. [PMID: 29893017 DOI: 10.1002/adma.201800316] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/27/2018] [Indexed: 05/16/2023]
Abstract
Glioblastoma is a most intractable and high-mortality malignancy because of its extremely low drug accessibility resulting from the blood-brain barrier (BBB). Here, it is reported that angiopep-2-directed and redox-responsive virus-mimicking polymersomes (ANG-PS) (angiopep-2 is a peptide targeting to low-density lipoprotein receptor-related protein-1 (LRP-1)) can efficiently and selectively chaperone saporin (SAP), a highly potent natural protein toxin, to orthotopic human glioblastoma xenografts in nude mice. Unlike chemotherapeutics, free SAP has a low cytotoxicity. SAP-loaded ANG-PS displays, however, a striking antitumor activity (half-maximal inhibitory concentration, IC50 = 30.2 × 10-9 m) toward U-87 MG human glioblastoma cells in vitro as well as high BBB transcytosis and glioblastoma accumulation in vivo. The systemic administration of SAP-loaded ANG-PS to U-87 MG orthotopic human-glioblastoma-bearing mice brings about little side effects, effective tumor inhibition, and significantly improved survival rate. The protein toxins chaperoned by LRP-1-targeted virus-mimicking vesicles emerge as a novel and highly promising treatment modality for glioblastoma.
Collapse
Affiliation(s)
- Yu Jiang
- 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, P. R. China
| | - Weijing Yang
- 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, P. R. China
| | - Jian Zhang
- 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, P. R. China
| | - Fenghua Meng
- 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, P. R. 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, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
45
|
Wu W, Luo L, Wang Y, Wu Q, Dai HB, Li JS, Durkan C, Wang N, Wang GX. Endogenous pH-responsive nanoparticles with programmable size changes for targeted tumor therapy and imaging applications. Theranostics 2018; 8:3038-3058. [PMID: 29896301 PMCID: PMC5996358 DOI: 10.7150/thno.23459] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/06/2018] [Indexed: 12/20/2022] Open
Abstract
Nanotechnology-based antitumor drug delivery systems, known as nanocarriers, have demonstrated their efficacy in recent years. Typically, the size of the nanocarriers is around 100 nm. It is imperative to achieve an optimum size of these nanocarriers which must be designed uniquely for each type of delivery process. For pH-responsive nanocarriers with programmable size, changes in pH (~6.5 for tumor tissue, ~5.5 for endosomes, and ~5.0 for lysosomes) may serve as an endogenous stimulus improving the safety and therapeutic efficacy of antitumor drugs. This review focuses on current advanced pH-responsive nanocarriers with programmable size changes for anticancer drug delivery. In particular, pH-responsive mechanisms for nanocarrier retention at tumor sites, size reduction for penetrating into tumor parenchyma, escaping from endo/lysosomes, and swelling or disassembly for drug release will be highlighted. Additional trends and challenges of employing these nanocarriers in future clinical applications are also addressed.
Collapse
Affiliation(s)
- Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Li Luo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Qi Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Han-Bin Dai
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Jian-Shu Li
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Colm Durkan
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
| | - Gui-Xue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| |
Collapse
|
46
|
Yang W, Xia Y, Fang Y, Meng F, Zhang J, Cheng R, Deng C, Zhong Z. Selective Cell Penetrating Peptide-Functionalized Polymersomes Mediate Efficient and Targeted Delivery of Methotrexate Disodium to Human Lung Cancer In Vivo. Adv Healthc Mater 2018; 7:e1701135. [PMID: 29280317 DOI: 10.1002/adhm.201701135] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/01/2017] [Indexed: 12/17/2022]
Abstract
It is a long challenge to develop nanomedicines that simultaneously possess tumor cell selectivity and penetration functions. Here, it is reported that selective cell penetrating peptide (RLWMRWYSPRTRAYGC)-functionalized polymersomes (SCPP-PS) mediate efficient and targeted delivery of methotrexate disodium (MTX) to human lung cancer in vivo. SCPP-PS with an SCPP density of 18.7% is self-crosslinked, has a small size (63-65 nm), and high MTX loading (up to 19.4 wt%), shows selective uptake and fast penetration into A549 lung cancer cells, and efficiently releases MTX intracellularly. Interestingly, MTX-loaded SCPP-PS (MTX-SCPP-PS) displays much lower IC50 than those of MTX-PS and free MTX. Installing SCPP to polymersomes has no detrimental effect to their long blood circulation time but significantly increases drug accumulation in A549 tumor (5.3% injected dose per gram at 8 h post injection). Remarkably, SCPP-PS exhibits deep penetration in to A549 tumors. MTX-SCPP-PS completely inhibits tumor progression and significantly improves survival rates in mice bearing A549 lung tumor xenografts as compared to MTX-PS and free MTX groups (median survival time: 75 vs 45 and 38 d, respectively), without causing noticeable adverse effects. These results highlight that functionalization of nanomedicines with SCPP is a feasible strategy to achieve efficient and targeted tumor therapy.
Collapse
Affiliation(s)
- Weijing Yang
- 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 P. R. China
| | - Yifeng Xia
- 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 P. R. China
| | - Yuan Fang
- 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 P. R. China
| | - Fenghua Meng
- 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 P. R. China
| | - Jian Zhang
- 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 P. R. China
| | - Ru Cheng
- 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 P. R. 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; Soochow University; Suzhou 215123 P. R. 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; Soochow University; Suzhou 215123 P. R. China
| |
Collapse
|
47
|
Zhang P, Steinborn B, Lächelt U, Zahler S, Wagner E. Lipo-Oligomer Nanoformulations for Targeted Intracellular Protein Delivery. Biomacromolecules 2017. [DOI: 10.1021/acs.biomac.7b00666] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Peng Zhang
- Pharmaceutical Biotechnology,
Center for System-based Drug Research Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Benjamin Steinborn
- Pharmaceutical Biotechnology,
Center for System-based Drug Research Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Ulrich Lächelt
- Pharmaceutical Biotechnology,
Center for System-based Drug Research Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Stefan Zahler
- Pharmaceutical Biotechnology,
Center for System-based Drug Research Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology,
Center for System-based Drug Research Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany
| |
Collapse
|
48
|
Zhao Y, Li X, Zhao X, Yang Y, Li H, Zhou X, Yuan W. Asymmetrical Polymer Vesicles for Drug delivery and Other Applications. Front Pharmacol 2017; 8:374. [PMID: 28676761 PMCID: PMC5476746 DOI: 10.3389/fphar.2017.00374] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/30/2017] [Indexed: 11/28/2022] Open
Abstract
Scientists have been attracted by polymersomes as versatile drug delivery systems since the last two decades. Polymersomes have the potential to be versatile drug delivery systems because of their tunable membrane formulations, stabilities in vivo, various physicochemical properties, controlled release mechanisms, targeting abilities, and capacities to encapsulate a wide range of drugs and other molecules. Asymmetrical polymersomes are nano- to micro-sized polymeric capsules with asymmetrical membranes, which means, they have different outer and inner coronas so that they can exhibit better endocytosis rate and endosomal escape ability than other polymeric systems with symmetrical membranes. Hence, asymmetrical polymersomes are highly promising as self-assembled nano-delivery systems in the future for in vivo therapeutics delivery and diagnostic imaging applications. In this review, we prepared a summary about recent research progresses of asymmetrical polymersomes in the following aspects: synthesis, preparation, applications in drug delivery and others.
Collapse
Affiliation(s)
- Yi Zhao
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghai, China
| | - Xiaoming Li
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghai, China
| | - Xiaotian Zhao
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghai, China
| | - Yunqi Yang
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghai, China
| | - Hui Li
- School of Medicine, University of California, San FranciscoSan Francisco, CA, United States
| | - Xinbo Zhou
- Laboratory of Computer-Aided Drug Design and Discovery, Beijing Institute of Pharmacology and ToxicologyBeijing, China
| | - Weien Yuan
- School of Pharmacy, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
49
|
Reactive oxygen species activated nanoparticles with tumor acidity internalization for precise anticancer therapy. J Control Release 2017; 255:142-153. [DOI: 10.1016/j.jconrel.2017.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 03/19/2017] [Accepted: 04/03/2017] [Indexed: 01/11/2023]
|
50
|
Mohammadi M, Ramezani M, Abnous K, Alibolandi M. Biocompatible polymersomes-based cancer theranostics: Towards multifunctional nanomedicine. Int J Pharm 2017; 519:287-303. [DOI: 10.1016/j.ijpharm.2017.01.037] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 01/20/2023]
|