1
|
Ding J, Zhang X, Guo L, Xiong J, Zhang C, Du Z, Zhu L, Alifu N, Dong B. NIR Triggered Bionic Bilayer Membrane-Encapsulated Nanoparticles for Synergistic Photodynamic, Photothermal and Chemotherapy of Cervical Cancer. Int J Nanomedicine 2025; 20:141-159. [PMID: 39802385 PMCID: PMC11721149 DOI: 10.2147/ijn.s496982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Purpose A synergistic treatment strategy of phototherapy and chemotherapy has been shown to improve efficacy and offer unique advantages over monotherapy. The purpose of this study is to explore a new nanocarrier system with liposome as the inner membrane and erythrocyte membrane as the outer membrane, which aims to realize the leak-free load of phototherapy drug indocyanine green (ICG) and chemotherapy drug doxorubicin (DOX), prolong the circulation time in vivo and improve the therapeutic effect. Patients and Methods In this study, bilayer membrane-loaded ICG and DOX nanoparticles (RBC@ICG-DOX NPs) were prepared and characterized. For in vitro analysis, the biocompatibility and tumor inhibition properties of the nanoparticles were evaluated. For in vivo analysis, the antitumor properties of the nanoparticles were explored in a mouse subcutaneous tumor model. Results RBC@ICG-DOX NPs were successfully prepared with strong safety and good blood compatibility, which can effectively reduce drug leakage and prolong drug circulation time in the body. In vitro performance evaluation showed that RBC@ICG-DOX NPs obtained excellent photothermal conversion ability and well reactive oxygen generation performance under near-infrared laser irradiation. Both in vitro and in vivo experiments showed well phototherapy-chemotherapy effect of RBC@ICG-DOX NPs with low toxic side effects. Conclusion Drug delivery, imaging and tumor synergies were accomplished through combinatorial strategies as well as bilayer membrane encapsulation, opening up a new platform for the design of future tumor combination therapies.
Collapse
Affiliation(s)
- Jiayi Ding
- Institute of Public Health, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Xueliang Zhang
- State Key Laboratory of Pathogenesis Prevention and Treatment of High Incidence Diseases in Central Asia, School of Medical Engineering and Technology Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Le Guo
- Institute of Public Health, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Jiabao Xiong
- Second Clinical Medical College, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Chi Zhang
- Institute of Public Health, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Zhong Du
- Second Clinical Medical College, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Lijun Zhu
- Second Clinical Medical College, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Nuernisha Alifu
- Institute of Public Health, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
- State Key Laboratory of Pathogenesis Prevention and Treatment of High Incidence Diseases in Central Asia, School of Medical Engineering and Technology Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
- Second Clinical Medical College, Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
| | - Biao Dong
- State Key Laboratory of Pathogenesis Prevention and Treatment of High Incidence Diseases in Central Asia, School of Medical Engineering and Technology Xinjiang Medical University, Urumqi, 830011, People’s Republic of China
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun, 130012, People’s Republic of China
| |
Collapse
|
2
|
Couvreur P. [Nanomedicines for the treatment of serious diseases and dual research]. C R Biol 2024; 347:187-198. [PMID: 39535509 DOI: 10.5802/crbiol.168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
|
3
|
Gao L, Dai X, Wu Y, Wang Y, Cheng L, Yan LT. Self-Assembly at Curved Biointerfaces. ACS NANO 2024; 18:30184-30210. [PMID: 39453716 DOI: 10.1021/acsnano.4c09675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Most of the biological interfaces are curved. Understanding the organizational structures and interaction patterns at such curved biointerfaces is therefore crucial not only for deepening our comprehension of the principles that govern life processes but also for designing and developing targeted drugs aimed at diseased cells and tissues. Despite the considerable efforts dedicated to this area of research, our understanding of curved biological interfaces is still limited. Many aspects of these interfaces remain elusive, presenting both challenges and opportunities for further exploration. In this review, we summarize the structural characteristics of biological interfaces found in nature, the current research status of materials associated with curved biointerfaces, and the theoretical advancements achieved to date. Finally, we outline future trends and challenges in the theoretical and technological development of curved biointerfaces. By addressing these challenges, people could bridge the knowledge gap and unlock the full potential of curved biointerfaces for scientific and technological advancements, ultimately benefiting various fields and improving human health and well-being.
Collapse
Affiliation(s)
- Lijuan Gao
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Xiaobin Dai
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Yibo Wu
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Yuming Wang
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Linghe Cheng
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Li-Tang Yan
- State Key Laboratory of Chemical Engineering, Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
4
|
Chenab KK, Malektaj H, Nadinlooie AAR, Mohammadi S, Zamani-Meymian MR. Intertumoral and intratumoral barriers as approaches for drug delivery and theranostics to solid tumors using stimuli-responsive materials. Mikrochim Acta 2024; 191:541. [PMID: 39150483 DOI: 10.1007/s00604-024-06583-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
The solid tumors provide a series of biological barriers in cellular microenvironment for designing drug delivery methods based on advanced stimuli-responsive materials. These intertumoral and intratumoral barriers consist of perforated endotheliums, tumor cell crowding, vascularity, lymphatic drainage blocking effect, extracellular matrix (ECM) proteins, hypoxia, and acidosis. Triggering opportunities have been drawn for solid tumor therapies based on single and dual stimuli-responsive drug delivery systems (DDSs) that not only improved drug targeting in deeper sites of the tumor microenvironments, but also facilitated the antitumor drug release efficiency. Single and dual stimuli-responsive materials which are known for their lowest side effects can be categorized in 17 main groups which involve to internal and external stimuli anticancer drug carriers in proportion to microenvironments of targeted solid tumors. Development of such drug carriers can circumvent barriers in clinical trial studies based on their superior capabilities in penetrating into more inaccessible sites of the tumor tissues. In recent designs, key characteristics of these DDSs such as fast response to intracellular and extracellular factors, effective cytotoxicity with minimum side effect, efficient permeability, and rate and location of drug release have been discussed as core concerns of designing paradigms of these materials.
Collapse
Affiliation(s)
- Karim Khanmohammadi Chenab
- Department of Chemistry, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
- Department of Physics, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, 9220, Aalborg, Denmark
| | | | | | | |
Collapse
|
5
|
Liu J, Cabral H, Mi P. Nanocarriers address intracellular barriers for efficient drug delivery, overcoming drug resistance, subcellular targeting and controlled release. Adv Drug Deliv Rev 2024; 207:115239. [PMID: 38437916 DOI: 10.1016/j.addr.2024.115239] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024]
Abstract
The cellular barriers are major bottlenecks for bioactive compounds entering into cells to accomplish their biological functions, which limits their biomedical applications. Nanocarriers have demonstrated high potential and benefits for encapsulating bioactive compounds and efficiently delivering them into target cells by overcoming a cascade of intracellular barriers to achieve desirable therapeutic and diagnostic effects. In this review, we introduce the cellular barriers ahead of drug delivery and nanocarriers, as well as summarize recent advances and strategies of nanocarriers for increasing internalization with cells, promoting intracellular trafficking, overcoming drug resistance, targeting subcellular locations and controlled drug release. Lastly, the future perspectives of nanocarriers for intracellular drug delivery are discussed, which mainly focus on potential challenges and future directions. Our review presents an overview of intracellular drug delivery by nanocarriers, which may encourage the future development of nanocarriers for efficient and precision drug delivery into a wide range of cells and subcellular targets.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
6
|
Wang Y, Huo Y, Zhao C, Liu H, Shao Y, Zhu C, An L, Chen X, Chen Z. Engineered exosomes with enhanced stability and delivery efficiency for glioblastoma therapy. J Control Release 2024; 368:170-183. [PMID: 38382811 DOI: 10.1016/j.jconrel.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/04/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Due to the blood-brain barrier (BBB), the application of chemical drugs for glioblastoma treatment is severely limited. Recently, exosomes have been widely applied for drug delivery to the brain. However, the differences in brain targeting efficiency among exosomes derived from different cell sources, as well as the premature drug leakage during circulation, still limit the therapeutic efficacy. Here, we designed a functional oligopeptide-modified exosome loaded with doxorubicin (Pep2-Exos-DOX) for glioblastoma treatment. BV2 mouse microglial cell line was selected as the exosome source due to the favorable BBB penetration. To avoid drug release in the circulation, a redox-response oligopeptide was designed for incorporation into the membranes of exosomes to lock the drug during circulation. The enrichment of the drug in glioblastoma was confirmed. Pharmacodynamic evaluation showed Pep2-Exos-DOX possessed significant anti-cancer activity against glioblastoma as well as relative biosafety. This exosome-based drug delivery system modified with redox-response oligopeptides provides us a novel strategy for brain diseases treatment.
Collapse
Affiliation(s)
- Yutong Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yiming Huo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chunyuan Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Heng Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali 671000, China
| | - Yurou Shao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chenqi Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lan An
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiao Chen
- School of Biopharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Zhipeng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
7
|
Gao LX, Hao H, Yu YQ, Chen JL, Chen WQ, Gong ZD, Liu Y, Jiang FL. Protein Labeling Facilitates the Understanding of Protein Corona Formation via Fluorescence Resonance Energy Transfer and Fluorescence Correlation Spectroscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:15275-15284. [PMID: 37853521 DOI: 10.1021/acs.langmuir.3c01986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Once nanoparticles enter into the biological milieu, nanoparticle-biomacromolecule complexes, especially the protein corona, swiftly form, which cause obvious effects on the physicochemical properties of both nanoparticles and proteins. Here, the thermodynamic parameters of the interactions between water-soluble GSH-CdSe/ZnS core/shell quantum dots (GSH-QDs) and human serum albumin (HSA) were investigated with the aid of labeling fluorescence of HSA. It was proved that the labeling fluorescence originating from a fluorophore (BDP-CN for instance) could be used to investigate the interactions between QDs and HSA. Gel electrophoresis displayed that the binding ratio between HSA and QDs was ∼2:1 by direct visualization. Fluorescence resonance energy transfer (FRET) results indicated that the distance between the QDs and the fluorophore BDP-CN in HSA was 7.2 nm, which indicated that the distance from the fluorophore to the surface of the QDs was ∼4.8 nm. Fluorescence correlation spectroscopy (FCS) results showed that HSA formed a monolayer of a protein corona with a thickness of 5.5 nm. According to the spatial structure of HSA, we could speculate that the binding site of QDs was located at the side edge (not the triangular plane) of HSA with an equilateral triangular prism. The elaboration of the thermodynamic parameters, binding ratio, and interaction orientation will highly improve the fundamental understanding of the formation of protein corona. This work has guiding significance for the exploration of the interactions between proteins and nanomaterials.
Collapse
Affiliation(s)
- Lian-Xun Gao
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Hao Hao
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Ying-Qi Yu
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Ji-Lei Chen
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Wen-Qi Chen
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Zuo-Dong Gong
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| | - Yi Liu
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, P. R. China
| | - Feng-Lei Jiang
- Sauvage Center for Molecular Sciences, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
8
|
Nandakumar A, Ito Y, Ueda M. Peptide-lipid hybrid vesicles with stimuli-responsive phase separation for controlled membrane functions. Chem Commun (Camb) 2023; 59:10644-10647. [PMID: 37580993 DOI: 10.1039/d3cc02954a] [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: 08/16/2023]
Abstract
A disulfide-tethered peptide-lipid conjugate self-assembled into a homogeneously distributed peptide-lipid hybrid vesicle. Upon dithiothreitol treatment, the homogeneous peptide-lipid membrane spontaneously divided into lipid-rich and peptide-rich domains, while the vesicle retained its size and shape. Membrane phase separation enhanced temperature-dependent cargo release.
Collapse
Affiliation(s)
- Avanashiappan Nandakumar
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science (CEMS), 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Motoki Ueda
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan.
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science (CEMS), 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| |
Collapse
|
9
|
Wang H, Qin L, Zhang X, Guan J, Mao S. Mechanisms and challenges of nanocarriers as non-viral vectors of therapeutic genes for enhanced pulmonary delivery. J Control Release 2022; 352:970-993. [PMID: 36372386 PMCID: PMC9671523 DOI: 10.1016/j.jconrel.2022.10.061] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022]
Abstract
With the rapid development of biopharmaceuticals and the outbreak of COVID-19, the world has ushered in a frenzy to develop gene therapy. Therefore, therapeutic genes have received enormous attention. However, due to the extreme instability and low intracellular gene expression of naked genes, specific vectors are required. Viral vectors are widely used attributed to their high transfection efficiency. However, due to the safety concerns of viral vectors, nanotechnology-based non-viral vectors have attracted extensive investigation. Still, issues of low transfection efficiency and poor tissue targeting of non-viral vectors need to be addressed. Especially, pulmonary gene delivery has obvious advantages for the treatment of inherited lung diseases, lung cancer, and viral pneumonia, which can not only enhance lung targeting and but also reduce enzymatic degradation. For systemic diseases therapy, pulmonary gene delivery can enhance vaccine efficacy via inducing not only cellular, humoral immunity but also mucosal immunity. This review provides a comprehensive overview of nanocarriers as non-viral vectors of therapeutic genes for enhanced pulmonary delivery. First of all, the characteristics and therapeutic mechanism of DNA, mRNA, and siRNA are provided. Thereafter, the advantages and challenges of pulmonary gene delivery in exerting local and systemic effects are discussed. Then, the inhalation dosage forms for nanoparticle-based drug delivery systems are introduced. Moreover, a series of materials used as nanocarriers for pulmonary gene delivery are presented, and the endosomal escape mechanisms of nanocarriers based on different materials are explored. The application of various non-viral vectors for pulmonary gene delivery are summarized in detail, with the perspectives of nano-vectors for pulmonary gene delivery.
Collapse
Affiliation(s)
| | | | - Xin Zhang
- Corresponding authors at: School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, China
| | | | - Shirui Mao
- Corresponding authors at: School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, 110016 Shenyang, China
| |
Collapse
|
10
|
Amin M, Lammers T, Ten Hagen TLM. Temperature-sensitive polymers to promote heat-triggered drug release from liposomes: Towards bypassing EPR. Adv Drug Deliv Rev 2022; 189:114503. [PMID: 35998827 DOI: 10.1016/j.addr.2022.114503] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/14/2022] [Accepted: 08/17/2022] [Indexed: 01/24/2023]
Abstract
Heat-triggered drug release from temperature-sensitive nanocarriers upon the application of mild hyperthermia is a promising approach to achieve site-specific delivery of drugs. The combination of mild hyperthermia (41-42 °C) and temperature-sensitive liposomes (TSL) that undergo lipid phase-transition and drug release has been studied extensively and has shown promising therapeutic outcome in a variety of animal tumor models as well as initial indications of success in humans. Sensitization of liposomes to mild hyperthermia by means of exploiting the thermal behavior of temperature-sensitive polymers (TSP) provides novel opportunities. Recently, TSP-modified liposomes (TSPL) have shown potential for enhancing tumor-directed drug delivery, either by triggered drug release or by triggered cell interactions in response to heat. In this review, we describe different classes of TSPL, and analyze and discuss the mechanisms and kinetics of content release from TSPL in response to local heating. In addition, the impact of lipid composition, polymer and copolymer characteristics, serum components and PEGylation on the mechanism of content release and TSPL performance is addressed. This is done from the perspective of rationally designing TSPL, with the overall goal of conceiving efficient strategies to increase the efficacy of TSPL plus hyperthermia to improve the outcome of targeted anticancer therapy.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Center for Biohybrid Medical Systems, Aachen, Germany.
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
11
|
Rasel MSI, Mohona FA, Akter W, Kabir S, Chowdhury AA, Chowdhury JA, Hassan MA, Al Mamun A, Ghose DK, Ahmad Z, Khan FS, Bari MF, Rahman MS, Amran MS. Exploration of Site-Specific Drug Targeting-A Review on EPR-, Stimuli-, Chemical-, and Receptor-Based Approaches as Potential Drug Targeting Methods in Cancer Treatment. JOURNAL OF ONCOLOGY 2022; 2022:9396760. [PMID: 36284633 PMCID: PMC9588330 DOI: 10.1155/2022/9396760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022]
Abstract
Cancer has been one of the most dominant causes of mortality globally over the last few decades. In cancer treatment, the selective targeting of tumor cells is indispensable, making it a better replacement for conventional chemotherapies by diminishing their adverse side effects. While designing a drug to be delivered selectively in the target organ, the drug development scientists should focus on various factors such as the type of cancer they are dealing with according to which drug, targeting moieties, and pharmaceutical carriers should be targeted. All published articles have been collected regarding cancer and drug-targeting approaches from well reputed databases including MEDLINE, Embase, Cochrane Library, CENTRAL and ClinicalTrials.gov, Science Direct, PubMed, Scopus, Wiley, and Springer. The articles published between January 2010 and December 2020 were considered. Due to the existence of various mechanisms, it is challenging to choose which one is appropriate for a specific case. Moreover, a combination of more than one approach is often utilized to achieve optimal drug effects. In this review, we have summarized and highlighted central mechanisms of how the targeted drug delivery system works in the specific diseased microenvironment, along with the strategies to make an approach more effective. We have also included some pictorial illustrations to have a precise idea about different types of drug targeting. The core contribution of this work includes providing a cancer drug development scientist with a broad preliminary idea to choose the appropriate approach among the various targeted drug delivery mechanisms. Also, the study will contribute to improving anticancer treatment approaches by providing a pathway for lesser side effects observed in conventional chemotherapeutic techniques.
Collapse
Affiliation(s)
- Md. Shamiul Islam Rasel
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Farhana Afrin Mohona
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Wahida Akter
- College of Pharmacy, University of Houston, Houston, USA
| | - Shaila Kabir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Abu Asad Chowdhury
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Jakir Ahmed Chowdhury
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| | - Md. Abul Hassan
- Department of Science & Technology, Tokushima University Graduate School, Tokushima, Japan
| | - Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang, China
| | - Dipayon Krisna Ghose
- Department of Biochemistry and Molecular Biology, Jagannath University, Dhaka 1100, Bangladesh
| | - Zubair Ahmad
- Unit of Bee Research and Honey Production, King Khalid University, Abha 61413, Saudi Arabia
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Department of Biology, College of Arts and Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Md. Fazlul Bari
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
| | - Md. Shah Amran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
| |
Collapse
|
12
|
Chiang PH, Fan CH, Jin Q, Yeh CK. Enhancing Doxorubicin Delivery in Solid Tumor by Superhydrophobic Amorphous Calcium Carbonate-Doxorubicin Silica Nanoparticles with Focused Ultrasound. Mol Pharm 2022; 19:3894-3905. [PMID: 36018041 DOI: 10.1021/acs.molpharmaceut.2c00384] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The current approach of delivering chemotherapy via pH-sensitive amorphous calcium carbonate-doxorubicin silica nanoparticles (ADS NPs) faces the challenge of insufficient drug dose due to drug instability within the bloodstream and poor tumor penetration. To overcome these long-standing obstacles, we proposed a superhydrophobic coating on the surface of the ADS NPs that could be easily modified via fluorination (ADSF NPs). The surface of fluorinated ADS NPs was further modified with a phospholipid layer to reduce aggregation and improve biocompatibility (ADSFL NPs). The contact angle and mean size of ADSFL NPs were 30.2 ± 4.4° and 353.1 ± 54.2 nm, respectively. The superhydrophobic layer generated interfacial nanobubbles on the outer shell of the NPs that reduced water-induced leakage of doxorubicin (DOX) sevenfold compared with the uncoated group and induced a cavitation effect upon ultrasound (US) sonication. Moreover, release of DOX from the ADSFL NPs could be triggered by US, and this release was further improved 1.6-fold in acidic aqueous conditions, indicating that the ADSFL NPs retained pH responsiveness. Enhanced sonography contrast and histological examination demonstrated that US could trigger cavitation activities from ADSFL NPs in vivo to induce vessel disruption and enhance the fluorescence intensity of DOX within the tumor region threefold under US imaging guidance compared with the ADSFL NPs-only group.
Collapse
Affiliation(s)
- Pei-Hua Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 30013, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, No. 1, University Road, Tainan 701, Taiwan.,Medical Device Innovation Center, National Cheng Kung University, No. 1, University Road, Tainan 701, Taiwan
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Avenue, Wuhan 430022, Hubei, China
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 30013, Taiwan
| |
Collapse
|
13
|
Asgari S, Ziarani GM, Badiei A, Pourjavadi A, Kiani M. A smart tri-layered nanofibrous hydrogel thin film with controlled release of dual drugs for chemo-thermal therapy of breast cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Li YL, Chen CY. Near-Infrared Light-Remote Localized Drug Delivery Systems Based on Zwitterionic Polymer Nanofibers for Combination Therapy. Polymers (Basel) 2022; 14:polym14091860. [PMID: 35567029 PMCID: PMC9100176 DOI: 10.3390/polym14091860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
Localized drug delivery systems (LDDS) have gained great interests because they can directly treat the tumors and minimize systematic toxicity, and maximize drug action by controlling release precisely at the tumor site. However, the resistance of the non-specific adsorption of biomolecules is also important to alleviate the inflammatory reactions and avoid the decrease in performance of LDDS. In this study, we develop a near infrared (NIR) light-triggered nanofibrous delivery system consisting of zwitterionic poly(2-methacryloyloxyethyl phosphorylcholine)-b-poly(ε-caprolactone) (PMPC-b-PCL) encapsulated with indocyanine green (ICG) and doxorubicin (DOX) for dual photothermal therapy and chemotherapy. The nanofibrous mat shows hydrophilic characteristics and good antifouling performance. Under mild NIR irradiation, ICG could convert NIR light into thermal energy that elevates the surrounding temperature above 45 °C. This thermal energy also markedly accelerates the DOX release from the nanofibrous mat due to softening of the nanofibers, indicating the drug release could be controlled and switched on/off by light-triggering. Moreover, this light-triggered thermal energy and releasing behavior contribute to enhancing the cell lethality. Intracellular DOX distribution confirms the more drugs release upon light irradiation. All results demonstrate the developed light-triggered drug release nanofibers as LDDS are biocompatible and antifouling as well as has the superior combinational chemotherapy/photothermal therapy.
Collapse
Affiliation(s)
| | - Ching-Yi Chen
- Correspondence: ; Tel.: +886-5-272-0411 (ext. 33409)
| |
Collapse
|
15
|
Veloso SRS, Tiryaki E, Spuch C, Hilliou L, Amorim CO, Amaral VS, Coutinho PJG, Ferreira PMT, Salgueiriño V, Correa-Duarte MA, Castanheira EMS. Tuning the drug multimodal release through a co-assembly strategy based on magnetic gels. NANOSCALE 2022; 14:5488-5500. [PMID: 35332904 DOI: 10.1039/d1nr08158f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Self-assembled short peptide-based gels are highly promising drug delivery systems. However, implementing a stimulus often requires screening different structures to obtain gels with suitable properties, and drugs might not be well encapsulated and/or cause undesirable effects on the gel's properties. To overcome this challenge, a new design approach is presented to modulate the release of doxorubicin as a model chemotherapeutic drug through the interplay of (di)phenylalanine-coated magnetic nanoparticles, PEGylated liposomes and doxorubicin co-assembly in dehydropeptide-based gels. The composites enable an enhancement of the gelation kinetics in a concentration-dependent manner, mainly through the use of PEGylated liposomes. The effect of the co-assembly of phenylalanine-coated nanoparticles with the hydrogel displays a concentration and size dependence. Finally, the integration of liposomes as doxorubicin storage units and of nanoparticles as composites that co-assemble with the gel matrix enables the tuneability of both passive and active doxorubicin release through a thermal, and a low-frequency alternating magnetic field-based trigger. In addition to the modulation of the gel properties, the functionalization with (di)phenylalanine improves the cytocompatibility of the nanoparticles. Hereby, this work paves a way for the development of peptide-based supramolecular systems for on-demand and controlled release of drugs.
Collapse
Affiliation(s)
- Sérgio R S Veloso
- Physics Centre of Minho and Porto Universities (CF-UM-UP) and LaPMET (Laboratory of Physics for Materials and Emergent Technologies), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| | - Ecem Tiryaki
- Departamento de Física Aplicada, Universidade de Vigo, 36310 Vigo, Spain
| | - Carlos Spuch
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, CIBERSAM, Hospital Álvaro Cunqueiro, Bloque Técnico, Planta 2, Sala de Investigación, Estrada Clara Campoamor, 341, 36212 Vigo, Spain
| | - Loic Hilliou
- Institute for Polymers and Composites, Department of Polymer Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal
| | - C O Amorim
- Physics Department and CICECO, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - V S Amaral
- Physics Department and CICECO, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Paulo J G Coutinho
- Physics Centre of Minho and Porto Universities (CF-UM-UP) and LaPMET (Laboratory of Physics for Materials and Emergent Technologies), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| | - Paula M T Ferreira
- Centro de Química (CQUM), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Verónica Salgueiriño
- Departamento de Física Aplicada, Universidade de Vigo, 36310 Vigo, Spain
- CINBIO, Universidad de Vigo, 36310 Vigo, Spain.
| | | | - Elisabete M S Castanheira
- Physics Centre of Minho and Porto Universities (CF-UM-UP) and LaPMET (Laboratory of Physics for Materials and Emergent Technologies), University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| |
Collapse
|
16
|
Samec T, Boulos J, Gilmore S, Hazelton A, Alexander-Bryant A. Peptide-based delivery of therapeutics in cancer treatment. Mater Today Bio 2022; 14:100248. [PMID: 35434595 PMCID: PMC9010702 DOI: 10.1016/j.mtbio.2022.100248] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 11/09/2022] Open
Abstract
Current delivery strategies for cancer therapeutics commonly cause significant systemic side effects due to required high doses of therapeutic, inefficient cellular uptake of drug, and poor cell selectivity. Peptide-based delivery systems have shown the ability to alleviate these issues and can significantly enhance therapeutic loading, delivery, and cancer targetability. Peptide systems can be tailor-made for specific cancer applications. This review describes three peptide classes, targeting, cell penetrating, and fusogenic peptides, as stand-alone nanoparticle systems, conjugations to nanoparticle systems, or as the therapeutic modality. Peptide nanoparticle design, characteristics, and applications are discussed as well as peptide applications in the clinical space.
Collapse
Affiliation(s)
- Timothy Samec
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Jessica Boulos
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Serena Gilmore
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Anthony Hazelton
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Angela Alexander-Bryant
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| |
Collapse
|
17
|
Nature-inspired dynamic gene-loaded nanoassemblies for the treatment of brain diseases. Adv Drug Deliv Rev 2022; 180:114029. [PMID: 34752841 DOI: 10.1016/j.addr.2021.114029] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 09/03/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022]
Abstract
Gene therapy has great potential to treat brain diseases. However, genetic drugs need to overcome a cascade of barriers for their full potential. The conventional delivery systems often struggle to meet expectations. Natural biological particles that are highly optimized for specific functions in body, can inspire optimization of dynamic gene-loaded nanoassemblies (DGN). The DGN refer to gene loaded nanoassemblies whose functions and structures are changeable in response to the biological microenvironments or can dynamically interact with tissues or cells. The nature-inspired DGN can meet the needs in brain diseases treatment, including i) Non-elimination in blood (N), ii) Across the blood-brain barrier (A), iii) Targeting cells (T), iv) Efficient uptake (U), v) Controllable release (R), vi) Eyeable (E)-abbreviated as the "NATURE". In this Review, from nature to "NATURE", we mainly summarize the specific application of nature-inspired DGN in the "NATURE" cascade process. Furthermore, the Review provides an outlook for this field.
Collapse
|
18
|
Xiang J, Liu X, Yuan G, Zhang R, Zhou Q, Xie T, Shen Y. Nanomedicine from amphiphilizedprodrugs: Concept and clinical translation. Adv Drug Deliv Rev 2021; 179:114027. [PMID: 34732344 DOI: 10.1016/j.addr.2021.114027] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/30/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
Nanomedicines generally consisting of carrier materials with small fractions of active pharmaceutical ingredients (API) have long been used to improve the pharmacokinetics and biodistributions, augment the therapeutic efficacies and mitigate the side effects. Amphiphilizing hydrophobic/hydrophilic drugs to prodrugs capable of self-assembly into well-defined nanostructures has emerged as a facile approach to fabricating nanomedicines because this amphiphilized prodrug (APD) strategy presents many advantages, including minimized use of inert carrier materials, well-characterized prodrug structures, fixed and high drug loading contents, 100% loading efficiency, and burst-free but controlled drug release. This review comprehensively summarizes recent advances in APDs and their nanomedicines, from the rationale and the stimuli-responsive linker chemistry for on-demand drug release to their progress to the clinics, clinical performance of APDs, as well as the challenges and perspective on future development.
Collapse
|
19
|
Lin YC, Fang TY, Kao HY, Tseng WC. Nanoassembly of UCST polypeptide for NIR-modulated drug release. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2021.108194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
A dual-responsive drug delivery system based on mesoporous silica nanoparticles covered with zipper-type peptide for intracellular transport/release. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
21
|
Štacko P, Šolomek T. Photoremovable Protecting Groups: Across the Light Spectrum to Near- Infrared Absorbing Photocages. Chimia (Aarau) 2021; 75:873-881. [PMID: 34728015 DOI: 10.2533/chimia.2021.873] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We discuss the past decade of progress in the field of photoremovable protecting groups that allowed the development of photocages activatable by near-IR light and highlight the individual conceptual advancements that lead to general guidelines to design new such photoremovable protecting groups. We emphasize the importance of understanding the individual photochemical reaction mechanisms that was necessary to achieve this progress and provide an outlook of the subsequent steps to facilitate a swift translation of this research into clinical praxis. Since this issue of CHIMIA is dedicated to the late Prof. Thomas Bally, we decided to provide a personal perspective on the field to which he contributed himself. We tried to write this review with the general readership of CHIMIA in mind in a hope to pay a tribute to the extraordinary dedication and clarity with which Thomas Bally used to explain abstract chemical concepts to his students or colleagues. We are uncertain whether we matched such challenge but we believe that he would have liked such approach very much.
Collapse
Affiliation(s)
- Peter Štacko
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich; Prievidza Chemical Society, M. Hodžu 10/16, 971 01 Prievidza, Slovakia;,
| | - Tomáš Šolomek
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland; Prievidza Chemical Society, M. Hodžu 10/16, 971 01 Prievidza, Slovakia;,
| |
Collapse
|
22
|
Zhang QW, Baig MMFA, Zhang TQ, Zhai TT, Qin X, Xia XH. RETRACTED: Liposomal valinomycin mediated cellular K + leak promoting apoptosis of liver cancer cells. J Control Release 2021; 337:317-328. [PMID: 34311027 DOI: 10.1016/j.jconrel.2021.07.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 01/18/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the corresponding author. It has been found that Fig 2B contains manipulated components, and Fig 5A partially overlaps with Fig 6 of a published paper authored by Mirza Muhammad Faran Ashraf Baig, et, al., The effective transfection of a low dose of negatively charged drug-loaded DNA-nanocarriers into cancer cells via scavenger receptors, J. Pharm. Anal. 11 (2021) 174-182, https://doi.org/10.1016/j.jpha.2020.10.003. The corresponding author indicated that they cannot guarantee the integrity of the images in the manuscript, as well as the conclusions of the paper. As a result, the Editor-in-Chief has decided to retract the paper. The corresponding author deeply regrets the circumstances and apologizes to the scientific community for not having detected this prior to publication.
Collapse
Affiliation(s)
- Qian-Wen Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Tian-Qi Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ting-Ting Zhai
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiang Qin
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xing-Hua Xia
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
23
|
Tavassoly O, Tavassoly I. Pharmacological Functionalization of Protein-Based Nanorobots as a Novel Tool for Drug Delivery in Cancer. ACS Pharmacol Transl Sci 2021; 4:1463-1467. [PMID: 34423277 DOI: 10.1021/acsptsci.1c00128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Indexed: 11/29/2022]
Abstract
The delivery of hydrophobic therapeutic agents to tumors is a challenge in the treatment of cancers. Here, we review recent advances in coiled-coil protein origami and discuss a proposed programmable protein origami structure, switchable by a protein kinase A/phosphatase switch, as an example of functionalization for designing future protein nanorobots.
Collapse
Affiliation(s)
- Omid Tavassoly
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Iman Tavassoly
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
24
|
Kiamohammadi L, Asadi L, Shirvalilou S, Khoei S, Khoee S, Soleymani M, Minaei SE. Physical and Biological Properties of 5-Fluorouracil Polymer-Coated Magnetite Nanographene Oxide as a New Thermosensitizer for Alternative Magnetic Hyperthermia and a Magnetic Resonance Imaging Contrast Agent: In Vitro and In Vivo Study. ACS OMEGA 2021; 6:20192-20204. [PMID: 34395970 PMCID: PMC8358959 DOI: 10.1021/acsomega.1c01763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/15/2021] [Indexed: 05/13/2023]
Abstract
This study reports a new procedure for utilizing 5-fluorouracil (5-Fu)-loaded polycaprolactone (PCL)/chitosan-covered magnetite nanographene oxide (5-Fu/SPION/NGO@PCL-LMWC) as a platform for synergistic thermo-chemotherapy. In fact, superparamagnetic iron oxide nanoparticles/nanographene oxide (SPION/NGO) nanoparticles can be coated with copolymers PCL/chitosan to attain better colloidal stability in the biological environment. Nanoparticles were synthesized and characterized for their size, surface charge, X-ray patterns, polymer content, and in vitro heat-triggered release. In vitro cytotoxic effects of nanoparticles on CT-26 cells were assessed with an MTT assay and real-time polymerase chain reaction. In vivo tumor growth inhibition was evaluated on an allograft mouse model of CT-26 cells. Tumor-bearing mice were injected with 5-Fu-loaded nanoparticles intravenously, and then, the targeted delivery was amplified using a magnetic field and finally exposed to an alternating magnetic field (AMF) (40 A/m, 13.56 MHz), during which the tumor site temperature increased to 43 °C. By using an infrared camera, we managed to heat the nanoparticles up to a constant temperature between 42.5 and 43.5 °C, with a tolerance ±0.03 °C. Finally, in vitro results showed that 5-Fu-loaded nanoparticles combined with AMF hyperthermia significantly reduced the plating efficiency of the cells (P < 0.01) and increased the Bax/Bcl-2 ratio (1.42 times, P < 0.01) compared with those achieved with each one alone. Furthermore, in vivo results demonstrated that the treatment of 5-Fu-loaded nanoparticles combined with the AMF diminished the growth of CT-26 tumor cells and increased the life span of the tumor-bearing mice (P < 0.001) by thermal energy deposition compared to that of the free 5-Fu drug. Also, the high level of accumulation of the nanoparticles within the tumor site was easily monitored with magnetic resonance imaging. It was concluded that the multifunctional magnetic nanoparticles could be used as a promising nanocarrier platform for achieving concurrent goals, drug delivery, magnetic targeting, thermal-sensitizing, cell death induction, and real-time monitoring of response to treatment.
Collapse
Affiliation(s)
- Leila Kiamohammadi
- Department
of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Leili Asadi
- Department
of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Sakine Shirvalilou
- Finetech
in Medicine Research Centre, Iran University
of Medical Sciences, Tehran 1449614535, Iran
| | - Samideh Khoei
- Finetech
in Medicine Research Centre, Iran University
of Medical Sciences, Tehran 1449614535, Iran
- Department
of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- , . Phone: (0098) 21 88622647. Fax: (0098) 21 88622647
| | - Sepideh Khoee
- Department
of Polymer Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran 14155 6455, Iran
| | - Maryam Soleymani
- Department
of Polymer Chemistry, School of Chemistry, College of Science, University of Tehran, Tehran 14155 6455, Iran
| | | |
Collapse
|
25
|
Luan X, Pan Y, Gao Y, Song Y. Recent near-infrared light-activated nanomedicine toward precision cancer therapy. J Mater Chem B 2021; 9:7076-7099. [PMID: 34124735 DOI: 10.1039/d1tb00671a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Light has been present throughout the history of mankind and even the universe. It is of great significance to human life, contributing to energy, agriculture, communication, and much more. In the biomedical field, light has been developed as a switch to control medical processes with minimal invasion and high spatiotemporal selectivity. During the past three years, near-infrared (NIR) light as long-wavelength light has been applied to more than 3000 achievements in biological applications due to its deep penetration depth and low phototoxicity. Remotely controlled cancer therapy usually involves the conversion of biologically inert NIR light. Thus, various materials, especially nanomaterials that can generate reactive oxygen species (ROS), ultraviolet (UV)/visual light, or thermal energy and so on under NIR illumination achieve great potential for the research of nanomedicine. Here, we offered an overview of recent advances in NIR light-activated nanomedicine for cancer therapeutic applications. NIR-light-conversion nanotechnologies for both directly triggering nanodrugs and smart drug delivery toward tumor therapy were discussed emphatically. The challenges and future trends of the use of NIR light in biomedical applications were also provided as a conclusion. We expect that this review will spark inspiration for biologists, materials scientists, pharmacologists, and chemists to fight against diseases and boost the future clinical-translational applications of NIR technology-based precision nanomedicine.
Collapse
Affiliation(s)
- Xiaowei Luan
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Advanced Icrostructures, Nanjing University, Nanjing, 210023, China.
| | - Yongchun Pan
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Advanced Icrostructures, Nanjing University, Nanjing, 210023, China.
| | - Yanfeng Gao
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Advanced Icrostructures, Nanjing University, Nanjing, 210023, China.
| | - Yujun Song
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, Collaborative Innovation Center of Advanced Icrostructures, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
26
|
Sun Y, Davis E. Nanoplatforms for Targeted Stimuli-Responsive Drug Delivery: A Review of Platform Materials and Stimuli-Responsive Release and Targeting Mechanisms. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:746. [PMID: 33809633 PMCID: PMC8000772 DOI: 10.3390/nano11030746] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
To achieve the promise of stimuli-responsive drug delivery systems for the treatment of cancer, they should (1) avoid premature clearance; (2) accumulate in tumors and undergo endocytosis by cancer cells; and (3) exhibit appropriate stimuli-responsive release of the payload. It is challenging to address all of these requirements simultaneously. However, the numerous proof-of-concept studies addressing one or more of these requirements reported every year have dramatically expanded the toolbox available for the design of drug delivery systems. This review highlights recent advances in the targeting and stimuli-responsiveness of drug delivery systems. It begins with a discussion of nanocarrier types and an overview of the factors influencing nanocarrier biodistribution. On-demand release strategies and their application to each type of nanocarrier are reviewed, including both endogenous and exogenous stimuli. Recent developments in stimuli-responsive targeting strategies are also discussed. The remaining challenges and prospective solutions in the field are discussed throughout the review, which is intended to assist researchers in overcoming interdisciplinary knowledge barriers and increase the speed of development. This review presents a nanocarrier-based drug delivery systems toolbox that enables the application of techniques across platforms and inspires researchers with interdisciplinary information to boost the development of multifunctional therapeutic nanoplatforms for cancer therapy.
Collapse
Affiliation(s)
| | - Edward Davis
- Materials Engineering Program, Mechanical Engineering Department, Auburn University, 101 Wilmore Drive, Auburn, AL 36830, USA;
| |
Collapse
|
27
|
Utterström J, Naeimipour S, Selegård R, Aili D. Coiled coil-based therapeutics and drug delivery systems. Adv Drug Deliv Rev 2021; 170:26-43. [PMID: 33378707 DOI: 10.1016/j.addr.2020.12.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/20/2022]
Abstract
Coiled coils are characterized by an arrangement of two or more α-helices into a superhelix and one of few protein motifs where the sequence-to-structure relationship to a large extent have been decoded and understood. The abundance of both natural and de novo designed coil coils provides a rich molecular toolbox for self-assembly of elaborate bespoke molecular architectures, nanostructures, and materials. Leveraging on the numerous possibilities to tune both affinities and preferences for polypeptide oligomerization, coiled coils offer unique possibilities to design modular and dynamic assemblies that can respond in a predictable manner to biomolecular interactions and subtle physicochemical cues. In this review, strategies to use coiled coils in design of novel therapeutics and advanced drug delivery systems are discussed. The applications of coiled coils for generating drug carriers and vaccines, and various aspects of using coiled coils for controlling and triggering drug release, and for improving drug targeting and drug uptake are described. The plethora of innovative coiled coil-based molecular systems provide new knowledge and techniques for improving efficacy of existing drugs and can facilitate development of novel therapeutic strategies.
Collapse
|
28
|
Phan H, Taresco V, Penelle J, Couturaud B. Polymerisation-induced self-assembly (PISA) as a straightforward formulation strategy for stimuli-responsive drug delivery systems and biomaterials: recent advances. Biomater Sci 2021; 9:38-50. [PMID: 33179646 DOI: 10.1039/d0bm01406k] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Stimuli-responsive amphiphilic block copolymers have emerged as promising nanocarriers for enhancing site-specific and on-demand drug release in response to a range of stimuli such as pH, the presence of redox agents, and temperature. The formulation of amphiphilic block copolymers into polymeric drug-loaded nanoparticles is typically achieved by various methods (e.g. oil-in-water emulsion solvent evaporation, solid dispersion, microphase separation, dialysis or microfluidic separation). Despite much progress that has been made, there remain many challenges to overcome to produce reliable polymeric systems. The main drawbacks of the above methods are that they produce very low solid contents (<1 wt%) and involve multiple-step procedures, thus limiting their scope. Recently, a new self-assembly methodology, polymerisation-induced self-assembly (PISA), has shown great promise in the production of polymer-derived particles using a straightforward one-pot approach, whilst facilitating high yield, scalability, and cost-effectiveness for pharmaceutical industry protocols. We therefore focus this review primarily on the most recent studies involved in the design and preparation of PISA-generated nano-objects which are responsive to specific stimuli, thus providing insight into how PISA may become an effective formulation strategy for the preparation of precisely tailored drug delivery systems and biomaterials, while some of the current challenges and limitations are also critically discussed.
Collapse
Affiliation(s)
- Hien Phan
- Univ Paris Est Creteil, CNRS, Institut de Chimie et des Matériaux Paris-Est (ICMPE), UMR 7182, 2 rue Henri Dunant, 94320 Thiais, France.
| | | | | | | |
Collapse
|
29
|
Roozbehi S, Dadashzadeh S, Sajedi RH. An enzyme-mediated controlled release system for curcumin based on cyclodextrin/cyclodextrin degrading enzyme. Enzyme Microb Technol 2020; 144:109727. [PMID: 33541570 DOI: 10.1016/j.enzmictec.2020.109727] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/05/2020] [Accepted: 12/14/2020] [Indexed: 12/28/2022]
Abstract
In this study, an enzyme-triggered system based on β-cyclodextrin (β-CD) has been developed to achieve controlled release of hydrophobic drugs in the presence of maltogenic amylase (MAase). The inclusion complex formation of curcumin (CUR), as a model anticancer compound, with β-CD was characterized by fluorescence and Fourier transform infrared (FTIR) spectroscopy. CUR was loaded into β-CD with an encapsulation efficiency of approximately 30 %. The in vitro profiles of CUR release from β-CD showed that 100 % of the drug was released after one hour incubation in the presence of MAase with cyclodextrin degrading activity. Fluorescence microscopy images indicate a significantly greater cellular uptake of CUR using β-CD-CUR/MAase system compared to β-CD-CUR inclusion complex without MAase. The β-CD-CUR/MAase system exhibited lower IC50 values and greater anti-proliferative effects in comparison with free CUR and β-CD-CUR in MCF-7 and Huh-7 cancer cells. The results from fluorescence microscopy and flow cytometric assay using the acridine orange/ethidium bromide and Annexin V-PE/7-AAD staining suggest that the β-CD-CUR/MAase system exhibited higher cytotoxic and apoptotic effects on cancer cells compared to other formulations. This triggered release of CUR in the presence of MAase is owing to the β-CD degradation by MAase resulting ring opening and chain scission in β-CD. We demonstrate that this enzyme-mediated controlled release system has a potential application for controlled release of poorly water-soluble drugs or hydrophobic compounds such as CUR.
Collapse
Affiliation(s)
- Sahar Roozbehi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran
| | - Simin Dadashzadeh
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Vali-e-asr Ave., Niayesh Junction, PO Box: 14155-6153, Tehran, Iran
| | - Reza H Sajedi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran.
| |
Collapse
|
30
|
Pérez-López A, Martín-Sabroso C, Torres-Suárez AI, Aparicio-Blanco J. Timeline of Translational Formulation Technologies for Cancer Therapy: Successes, Failures, and Lessons Learned Therefrom. Pharmaceutics 2020; 12:E1028. [PMID: 33126622 PMCID: PMC7692572 DOI: 10.3390/pharmaceutics12111028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past few decades, the field of cancer therapy has seen a significant change in the way in which formulations are designed and developed, resulting in more efficient products that allow us to ultimately achieve improved drug bioavailability, efficacy, and safety. However, although many formulations have entered the market, many others have fallen by the wayside leaving the scientific community with several lessons to learn. The successes (and failures) achieved with formulations that have been approved in Europe and/or by the FDA for the three major types of cancer therapy (peptide-based therapy, chemotherapy, and radiotherapy) are reviewed herein, covering the period from the approval of the first prolonged-release system for hormonal therapy to the appearance of the first biodegradable microspheres intended for chemoembolization in 2020. In addition, those products that have entered phase III clinical trials that have been active over the last five years are summarized in order to outline future research trends and possibilities that lie ahead to develop clinically translatable formulations for cancer treatment.
Collapse
Affiliation(s)
- Alexandre Pérez-López
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.P.-L.); (C.M.-S.); (J.A.-B.)
| | - Cristina Martín-Sabroso
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.P.-L.); (C.M.-S.); (J.A.-B.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.P.-L.); (C.M.-S.); (J.A.-B.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Juan Aparicio-Blanco
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.P.-L.); (C.M.-S.); (J.A.-B.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
31
|
Amin M, Huang W, Seynhaeve ALB, ten Hagen TLM. Hyperthermia and Temperature-Sensitive Nanomaterials for Spatiotemporal Drug Delivery to Solid Tumors. Pharmaceutics 2020; 12:E1007. [PMID: 33105816 PMCID: PMC7690578 DOI: 10.3390/pharmaceutics12111007] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Nanotechnology has great capability in formulation, reduction of side effects, and enhancing pharmacokinetics of chemotherapeutics by designing stable or long circulating nano-carriers. However, effective drug delivery at the cellular level by means of such carriers is still unsatisfactory. One promising approach is using spatiotemporal drug release by means of nanoparticles with the capacity for content release triggered by internal or external stimuli. Among different stimuli, interests for application of external heat, hyperthermia, is growing. Advanced technology, ease of application and most importantly high level of control over applied heat, and as a result triggered release, and the adjuvant effect of hyperthermia in enhancing therapeutic response of chemotherapeutics, i.e., thermochemotherapy, make hyperthermia a great stimulus for triggered drug release. Therefore, a variety of temperature sensitive nano-carriers, lipid or/and polymeric based, have been fabricated and studied. Importantly, in order to achieve an efficient therapeutic outcome, and taking the advantages of thermochemotherapy into consideration, release characteristics from nano-carriers should fit with applicable clinical thermal setting. Here we introduce and discuss the application of the three most studied temperature sensitive nanoparticles with emphasis on release behavior and its importance regarding applicability and therapeutic potentials.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Wenqiu Huang
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
| | - Ann L. B. Seynhaeve
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
| | - Timo L. M. ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands; (M.A.); (W.H.); (A.L.B.S.)
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| |
Collapse
|
32
|
Condello M, Mancini G, Meschini S. The Exploitation of Liposomes in the Inhibition of Autophagy to Defeat Drug Resistance. Front Pharmacol 2020; 11:787. [PMID: 32547395 PMCID: PMC7272661 DOI: 10.3389/fphar.2020.00787] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a mechanism involved in many human diseases and in cancers can have a cytotoxic/cytostatic or protective action, being in the latter case involved in multidrug resistance. Understanding which of these roles autophagy has in cancer is thus fundamental for therapeutical decisions because it permits to optimize the therapeutical approach by activating or inhibiting autophagy according to the progression of the disease. However, a serious drawback of cancer treatment is often the scarce availability of drugs and autophagy modulators at the sites of interest. In the recent years, several nanocarriers have been developed and investigated to improve the solubility, bioavailability, controlled release of therapeutics and increase their cytotoxic effect on cancer cell. Here we have reviewed only liposomes as carriers of chemotherapeutics and autophagy inhibitors because they have low toxicity and immunogenicity and they are biodegradable and versatile. In this review after the analysis of the dual role of autophagy, of the main autophagic pathways, and of the role of autophagy in multidrug resistance, we will focus on the most effective liposomal formulations, thus highlighting the great potential of these targeting systems to defeat cancer diseases.
Collapse
Affiliation(s)
- Maria Condello
- National Center for Drug Research and Evaluation, National Institute of Health, Rome, Italy
| | - Giovanna Mancini
- Institute for Biological Systems, National Research Council, Rome, Italy
| | - Stefania Meschini
- National Center for Drug Research and Evaluation, National Institute of Health, Rome, Italy
| |
Collapse
|
33
|
Feger G, Angelov B, Angelova A. Prediction of Amphiphilic Cell-Penetrating Peptide Building Blocks from Protein-Derived Amino Acid Sequences for Engineering of Drug Delivery Nanoassemblies. J Phys Chem B 2020; 124:4069-4078. [PMID: 32337991 DOI: 10.1021/acs.jpcb.0c01618] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amphiphilic molecules, forming self-assembled nanoarchitectures, are typically composed of hydrophobic and hydrophilic domains. Peptide amphiphiles can be designed from two, three, or four building blocks imparting novel structural and functional properties and affinities for interaction with cellular membranes or intracellular organelles. Here we present a combined numerical approach to design amphiphilic peptide scaffolds that are derived from the human nuclear Ki-67 protein. Ki-67 acts, like a biosurfactant, as a steric and electrostatic charge barrier against the collapse of mitotic chromosomes. The proposed predictive design of new Ki-67 protein-derived amphiphilic amino acid sequences exploits the computational outcomes of a set of web-accessible predictors, which are based on machine learning methods. The ensemble of such artificial intelligence algorithms, involving support vector machine (SVM), random forest (RF) classifiers, and neural networks (NN), enables the nanoengineering of a broad range of innovative peptide materials for therapeutic delivery in various applications. Amphiphilic cell-penetrating peptides (CPP), derived from natural protein sequences, may spontaneously form self-assembled nanocarriers characterized by enhanced cellular uptake. Thanks to their inherent low immunogenicity, they may enable the safe delivery of therapeutic molecules across the biological barriers.
Collapse
Affiliation(s)
- Guillaume Feger
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay UMR8612, F-92296 Châtenay-Malabry, France
| | - Borislav Angelov
- Institute of Physics, ELI Beamlines, Academy of Sciences of the Czech Republic, Na Slovance 2, CZ-18221 Prague, Czech Republic
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay UMR8612, F-92296 Châtenay-Malabry, France
| |
Collapse
|
34
|
Saccardo A, Soloviev M, Ferrari E. A thermo-responsive, self-assembling biointerface for on demand release of surface-immobilised proteins. Biomater Sci 2020; 8:2673-2681. [PMID: 32254844 DOI: 10.1039/c9bm01957j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dedicated chemistries for on-demand capture and release of biomolecules at the solid-liquid interface are required for applications in drug delivery, for the synthesis of switchable surfaces used in analytical devices and for the assembly of next-generation biomaterials with complex architectures and functions. Here we report the engineering of a binary self-assembling polypeptide system for reversible protein capture, immobilisation and controlled thermo-responsive release from a solid surface. The first element of the binary system is a universal protein substrate immobilised on a solid surface. This protein is bio-inspired by the neuronal SNAP25, which is the protein involved in the docking and fusion of synaptic vesicles to the synaptic membrane. The second element is an artificial chimeric protein engineered to include distinct domains from three different proteins: Syntaxin, VAMP and SNAP25. These native proteins constitute the machinery dedicated to vesicle trafficking in eukaryotes. We removed approximately 70% of native protein sequence from these proteins and constructed a protein chimera capable of high affinity interaction and self-assembly with immobilised substrate. The interaction of the two parts of the engineered protein complex is strong but fully-reversible and therefore the chimera can be recombinantly fused as a tag to a protein of interest, to allow spontaneous assembly and stimuli-sensitive release from the surface upon heating at a predetermined temperature. Two thermo-responsive tags are reported: the first presents remarkable thermal stability with melting temperature of the order of 80 °C; the second disassembles at a substantially lower temperature of about 45 °C. The latter is a promising candidate for remote-controlled localised delivery of therapeutic proteins, as physiologically tolerable local increase of temperatures in the 40-45 °C range can be achieved using magnetic fields, infra-red light or focused ultrasound. Importantly, these two novel polypeptides provide a broader blueprint for the engineering of future functional proteins with predictable folding and response to external stimuli.
Collapse
Affiliation(s)
- Angela Saccardo
- School of Life Sciences, University of Lincoln, Lincoln LN6 7TS, UK.
| | - Mikhail Soloviev
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, UK
| | - Enrico Ferrari
- School of Life Sciences, University of Lincoln, Lincoln LN6 7TS, UK.
| |
Collapse
|
35
|
Abstract
Peptides are one of the most important functional motifs for constructing smart drug delivery systems (DDSs). Functional peptides can be conjugated with drugs or carriers via covalent bonds, or assembled into DDSs via supramolecular forces, which enables the DDSs to acquire desired functions such as targeting and/or environmental responsiveness. In this mini review, we first introduce the different types of functional peptides that are commonly used for constructing DDSs, and we highlight representative strategies for designing smart DDSs by using functional peptides in the past few years. We also state the challenges of peptide-based DDSs and come up with prospects.
Collapse
Affiliation(s)
- Zheng Lian
- People's Public Security University of China, Beijing 100038, China
| | | |
Collapse
|
36
|
Wang C, Ye X, Zhao Y, Bai L, He Z, Tong Q, Xie X, Zhu H, Cai D, Zhou Y, Lu B, Wei Y, Mei L, Xie D, Wang M. Cryogenic 3D printing of porous scaffolds for in situ delivery of 2D black phosphorus nanosheets, doxorubicin hydrochloride and osteogenic peptide for treating tumor resection-induced bone defects. Biofabrication 2020; 12:035004. [DOI: 10.1088/1758-5090/ab6d35] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
37
|
Mi P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020; 10:4557-4588. [PMID: 32292515 PMCID: PMC7150471 DOI: 10.7150/thno.38069] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/24/2020] [Indexed: 02/05/2023] Open
Abstract
In recent years, much progress has been motivated in stimuli-responsive nanocarriers, which could response to the intrinsic physicochemical and pathological factors in diseased regions to increase the specificity of drug delivery. Currently, numerous nanocarriers have been engineered with physicochemical changes in responding to external stimuli, such as ultrasound, thermal, light and magnetic field, as well as internal stimuli, including pH, redox potential, hypoxia and enzyme, etc. Nanocarriers could respond to stimuli in tumor microenvironments or inside cancer cells for on-demanded drug delivery and accumulation, controlled drug release, activation of bioactive compounds, probes and targeting ligands, as well as size, charge and conformation conversion, etc., leading to sensing and signaling, overcoming multidrug resistance, accurate diagnosis and precision therapy. This review has summarized the general strategies of developing stimuli-responsive nanocarriers and recent advances, presented their applications in drug delivery, tumor imaging, therapy and theranostics, illustrated the progress of clinical translation and made prospects.
Collapse
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, 610041, China
| |
Collapse
|
38
|
Pinto MN, Mascharak PK. Light-assisted and remote delivery of carbon monoxide to malignant cells and tissues: Photochemotherapy in the spotlight. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY C-PHOTOCHEMISTRY REVIEWS 2020. [DOI: 10.1016/j.jphotochemrev.2020.100341] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Comprehensive Effects of Near-Infrared Multifunctional Liposomes on Cancer Cells. Molecules 2020; 25:molecules25051098. [PMID: 32121482 PMCID: PMC7179136 DOI: 10.3390/molecules25051098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Multifunctional theranostic systems are a recent important development of medical research. We combined the characteristics of near-infrared luminescent quantum dots and thermosensitive magnetoliposomes to develop a multifunctional nano-diagnostic material. This system is based on near-infrared magnetic thermosensitive liposomes, which encapsulate drugs and can control drug localization and release. After incubating cancer cells with the liposomes, the state of the cells was analyzed in real time by near-infrared imaging. Cell viability was significantly inhibited by heat treatment or alternating magnetic field treatment, which thus improved the anti-cancer properties of the liposomes. In the future, by combining near-infrared imaging technology and an external high-frequency alternating magnetic field, we could not only detect cancer cells noninvasively but also conduct image-guided treatments for cancer.
Collapse
|
40
|
Zhang W, Yu W, Ding X, Yin C, Yan J, Yang E, Guo F, Sun D, Wang W. Self-assembled thermal gold nanorod-loaded thermosensitive liposome-encapsulated ganoderic acid for antibacterial and cancer photochemotherapy. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2019; 47:406-419. [PMID: 30724609 DOI: 10.1080/21691401.2018.1559177] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
Abstract
A novel nanoparticle (Au-LTSL-GA.A) uses the thermosensitive liposome (LTSL) to encapsulate ganoderic acid A (GA.A), which successfully transforms the polarity of GA.A and has excellent water solubility. The multifunctional Au-LTSL-GA.A, a self-assembled thermal nanomaterial, was used in antibacterial and anticancer applications in combination with near-infrared (NIR) irradiation. The designed Au-LTSL-GA.A nanoparticle was used as a nano-photosensitizer to achieve synergistic photochemotherapy based on the phototherapy sensitization property of Au nanorods (NRs) and antitumour activity of GA.A. In the antibacterial experiments, the Au-LTSL-GA.A + NIR irradiation had a broad-spectrum antibacterial effect, exhibiting a strong antibacterial activity against drug-resistant Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) compared with the raw GA.A and LTSL-GA.A. In the anticancer experiments, Au-LTSL-GA.A + NIR irradiation, which combined phototherapy sensitization property of Au NRs with antitumour activity of GA.A, exhibited high anticancer activity against MCF-7 cells. The IC50 value of Au-LTSL-GA.A + NIR irradiation (12.1 ± 1.3 μg/mL) was almost similar to cisplatin in MCF-7 cells. The evaluation of the potential in vivo toxicity of Au-LTSL-GA.A revealed no toxicity in mice. The results of this study suggest that Au-LTSL-GA.A has a wide range of potential industrial and clinical applications, such as in antibacterial treatment and cancer photochemotherapy.
Collapse
Affiliation(s)
- Weiwei Zhang
- a School of Life Sciences , Anhui Agricultural University , Hefei , China
| | - Wenwen Yu
- a School of Life Sciences , Anhui Agricultural University , Hefei , China
| | - Xiaoyuan Ding
- a School of Life Sciences , Anhui Agricultural University , Hefei , China
| | - Chenyang Yin
- a School of Life Sciences , Anhui Agricultural University , Hefei , China
| | - Jing Yan
- a School of Life Sciences , Anhui Agricultural University , Hefei , China
| | - Endong Yang
- a School of Life Sciences , Anhui Agricultural University , Hefei , China
| | - Feng Guo
- a School of Life Sciences , Anhui Agricultural University , Hefei , China
| | - Dongdong Sun
- a School of Life Sciences , Anhui Agricultural University , Hefei , China
| | - Weiyun Wang
- a School of Life Sciences , Anhui Agricultural University , Hefei , China
| |
Collapse
|
41
|
Mondal S, Basavalingappa V, Jacoby G, Shimon LJW, Beck R, Gazit E. Functional Coiled-Coil-like Assembly by Knob-into-Hole Packing of Single Heptad Repeat. ACS NANO 2019; 13:12630-12637. [PMID: 31647865 PMCID: PMC7616943 DOI: 10.1021/acsnano.9b04148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Coiled-coil peptides represent the principal building blocks for structure-based design of bionanomaterials. The sequence-structure relationship and precise nanoscale ordering of the coiled-coil helices originate from the knob-into-hole (KIH) packing of side chains. The helical interface stabilized by the KIH interaction is known to have chain lengths ranging from 30 to 1000 residues. Yet the shortest peptide required for oligomerization through KIH assembly is still unknown. Here, we report that through atomic resolution a minimal seven-residue amphipathic helix forms a different type of KIH motif, termed "supramolecular KIH packing", which confers an exceptional stability to the helical dimers. Significantly, at a low pH, the peptide self-assembles into nanofibers with coiled-coil architecture resembling the natural fibrous proteins. Furthermore, hierarchical ordering of the nanofibers affords lyotropic liquid crystals composed of a shortest natural helical sequence. Thus, this study expands the sequence space for a coiled-coil folding manifold and provides another paradigm for designer nanomaterials from minimal helical sequences.
Collapse
Affiliation(s)
- Sudipta Mondal
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Vasantha Basavalingappa
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Guy Jacoby
- The Raymond and Beverly Sackler School of Physics and Astronomy, Tel Aviv University, Tel Aviv 69978, Israel
| | - Linda J. W. Shimon
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Roy Beck
- The Raymond and Beverly Sackler School of Physics and Astronomy, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
42
|
Al-Ahmady ZS, Jasim D, Ahmad SS, Wong R, Haley M, Coutts G, Schiessl I, Allan SM, Kostarelos K. Selective Liposomal Transport through Blood Brain Barrier Disruption in Ischemic Stroke Reveals Two Distinct Therapeutic Opportunities. ACS NANO 2019; 13:12470-12486. [PMID: 31693858 DOI: 10.1021/acsnano.9b01808] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The development of effective therapies for stroke continues to face repeated translational failures. Brain endothelial cells form paracellular and transcellular barriers to many blood-borne therapies, and the development of efficient delivery strategies is highly warranted. Here, in a mouse model of stroke, we show selective recruitment of clinically used liposomes into the ischemic brain that correlates with biphasic blood brain barrier (BBB) breakdown. Intravenous administration of liposomes into mice exposed to transient middle cerebral artery occlusion took place at early (0.5 and 4 h) and delayed (24 and 48 h) time points, covering different phases of BBB disruption after stroke. Using a combination of in vivo real-time imaging and histological analysis we show that selective liposomal brain accumulation coincides with biphasic enhancement in transcellular transport followed by a delayed impairment to the paracellular barrier. This process precedes neurological damage in the acute phase and maintains long-term liposomal colocalization within the neurovascular unit, which could have great potential for neuroprotection. Levels of liposomal uptake by glial cells are similarly selectively enhanced in the ischemic region late after experimental stroke (2-3 days), highlighting their potential for blocking delayed inflammatory responses or shifting the polarization of microglia/macrophages toward brain repair. These findings demonstrate the capability of liposomes to maximize selective translocation into the brain after stroke and identify two windows for therapeutic manipulation. This emphasizes the benefits of selective drug delivery for efficient tailoring of stroke treatments.
Collapse
Affiliation(s)
- Zahraa S Al-Ahmady
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building , The University of Manchester , Manchester M13 9PT , United Kingdom
- Pharmacology Department, School of Science and Technology , Nottingham Trent University , Nottingham NG11 8NS , United Kingdom
| | - Dhifaf Jasim
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building , The University of Manchester , Manchester M13 9PT , United Kingdom
| | - Sabahuddin Syed Ahmad
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building , The University of Manchester , Manchester M13 9PT , United Kingdom
| | - Raymond Wong
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Manchester Academic Health Science Centre, AV Hill Building, Manchester M13 9PT , United Kingdom
| | - Michael Haley
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Manchester Academic Health Science Centre, AV Hill Building, Manchester M13 9PT , United Kingdom
| | - Graham Coutts
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Manchester Academic Health Science Centre, AV Hill Building, Manchester M13 9PT , United Kingdom
| | - Ingo Schiessl
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Manchester Academic Health Science Centre, AV Hill Building, Manchester M13 9PT , United Kingdom
| | - Stuart M Allan
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Manchester Academic Health Science Centre, AV Hill Building, Manchester M13 9PT , United Kingdom
| | - Kostas Kostarelos
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, AV Hill Building , The University of Manchester , Manchester M13 9PT , United Kingdom
| |
Collapse
|
43
|
Al-Ahmady ZS, Donno R, Gennari A, Prestat E, Marotta R, Mironov A, Newman L, Lawrence MJ, Tirelli N, Ashford M, Kostarelos K. Enhanced Intraliposomal Metallic Nanoparticle Payload Capacity Using Microfluidic-Assisted Self-Assembly. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:13318-13331. [PMID: 31478662 DOI: 10.1021/acs.langmuir.9b00579] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Hybrids composed of liposomes (L) and metallic nanoparticles (NPs) hold great potential for imaging and drug delivery purposes. However, the efficient incorporation of metallic NPs into liposomes using conventional methodologies has so far proved to be challenging. In this study, we report the fabrication of hybrids of liposomes and hydrophobic gold NPs of size 2-4 nm (Au) using a microfluidic-assisted self-assembly process. The incorporation of increasing amounts of AuNPs into liposomes was examined using microfluidics and compared to L-AuNP hybrids prepared by the reverse-phase evaporation method. Our microfluidics strategy produced L-AuNP hybrids with a homogeneous size distribution, a smaller polydispersity index, and a threefold increase in loading efficiency when compared to those hybrids prepared using the reverse-phase method of production. Quantification of the loading efficiency was determined by ultraviolet spectroscopy, inductively coupled plasma mass spectroscopy, and centrifugal field flow fractionation, and qualitative validation was confirmed by transmission electron microscopy. The higher loading of gold NPs into the liposomes achieved using microfluidics produced a slightly thicker and more rigid bilayer as determined with small-angle neutron scattering. These observations were confirmed using fluorescent anisotropy and atomic force microscopy. Structural characterization of the liposomal-NP hybrids with cryo-electron microscopy revealed the coexistence of membrane-embedded and interdigitated NP-rich domains, suggesting AuNP incorporation through hydrophobic interactions. The microfluidic technique that we describe in this study allows for the automated production of monodisperse liposomal-NP hybrids with high loading capacity, highlighting the utility of microfluidics to improve the payload of metallic NPs within liposomes, thereby enhancing their application for imaging and drug delivery.
Collapse
Affiliation(s)
- Zahraa S Al-Ahmady
- Nanomedicine Lab, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health , University of Manchester , Av Hill Building , Manchester M13 9PT , U.K
- Pharmacology Department, School of Science and Technology , Nottingham Trent University , Nottingham NG11 8NS , U.K
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
| | - Roberto Donno
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
- Laboratory of Polymers and Biomaterials , Fondazione Istituto Italiano di Tecnologia , 16163 , Genova , Italy
| | - Arianna Gennari
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
- Laboratory of Polymers and Biomaterials , Fondazione Istituto Italiano di Tecnologia , 16163 , Genova , Italy
| | - Eric Prestat
- SuperSTEM Laboratory , SciTech Daresbury Campus , Keckwick Lane, Warrington WA4 4AD , U.K
| | - Roberto Marotta
- Electron Microscopy Laboratory , Fondazione Istituto Italiano di Tecnologia , 16163 Genova , Italy
| | | | - Leon Newman
- Nanomedicine Lab, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health , University of Manchester , Av Hill Building , Manchester M13 9PT , U.K
| | - M Jayne Lawrence
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
| | - Nicola Tirelli
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
- Laboratory of Polymers and Biomaterials , Fondazione Istituto Italiano di Tecnologia , 16163 , Genova , Italy
| | - Marianne Ashford
- Advanced Drug Delivery Pharmaceutical Sciences, IMED Biotech Unit , AstraZeneca , Macclesfield SK10 2NA , U.K
| | - Kostas Kostarelos
- Nanomedicine Lab, Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health , University of Manchester , Av Hill Building , Manchester M13 9PT , U.K
- North West Centre of Advanced Drug Delivery (NoWCADD), Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health , University of Manchester , Stopford Building , Manchester , M13 9PT , U.K
| |
Collapse
|
44
|
Novel therapeutic interventions in cancer treatment using protein and peptide-based targeted smart systems. Semin Cancer Biol 2019; 69:249-267. [PMID: 31442570 DOI: 10.1016/j.semcancer.2019.08.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 01/12/2023]
Abstract
Cancer, being the most prevalent and resistant disease afflicting any gender, age or social status, is the ultimate challenge for the scientific community. The new generation therapeutics for cancer management has shifted the approach to personalized/precision medicine, making use of patient- and tumor-specific markers for specifying the targeted therapies for each patient. Peptides targeting these cancer-specific signatures hold enormous potential for cancer therapy and diagnosis. The rapid advancements in the combinatorial peptide libraries served as an impetus to the development of multifunctional peptide-based materials for targeted cancer therapy. The present review outlines benefits and shortcomings of peptides as cancer therapeutics and the potential of peptide modified nanomedicines for targeted delivery of anticancer agents.
Collapse
|
45
|
Two/three-dimensional interfacial properties of the novel peptide as a selective destroyer of biomembrane. Colloids Surf A Physicochem Eng Asp 2019. [DOI: 10.1016/j.colsurfa.2019.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
46
|
Zhang L, Shen S, Cheng L, You H, Lu L, Ma C, Dai Y, Fang J. Mesoporous gold nanoparticles for photothermal controlled anticancer drug delivery. Nanomedicine (Lond) 2019; 14:1443-1454. [DOI: 10.2217/nnm-2018-0242] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Aim: To realize the transit and release of cancer drug exactly as well as high drug loading ratio, we reported a biocompatible and temperature responsive controlled drug delivery system based on 3D mesoporous structured Au networks. Materials & methods: Here, we filled the hollow interiors of Au networks with a phase-change material so that the drug release was easily regulated by controlling the temperature only. Results: Thanks to the high near-infrared reflectance absorbance and mesoporous structure, the Au–PEG + lauric acid/doxorubicin system showed a strong photothermal conversion efficiency, high drug-loading ratio (54.2% for doxorubicin) and controlled drug release. Conclusion: This system revealed great advantages in photothermal therapy and chemotherapy, offering an obvious synergistic effect in cancer treatment.
Collapse
Affiliation(s)
- Lingling Zhang
- School of Electronic & Information Engineering, Xi’an Jiaotong University, Xi’an, Shann xi 710049, PR China
| | - Sida Shen
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Hongjun You
- School of Science, Xi’an Jiaotong University, Xi’an, Shann xi 710049, PR China
| | - Lu Lu
- School of Microelectronics, Xi’an Jiaotong University, Xi’an, Shann xi 710049, PR China
| | - Chuansheng Ma
- School of Microelectronics, Xi’an Jiaotong University, Xi’an, Shann xi 710049, PR China
| | - Yanzhu Dai
- School of Microelectronics, Xi’an Jiaotong University, Xi’an, Shann xi 710049, PR China
| | - Jixiang Fang
- School of Electronic & Information Engineering, Xi’an Jiaotong University, Xi’an, Shann xi 710049, PR China
| |
Collapse
|
47
|
Sarkar S, Dinda S, Choudhury P, Kumar Das P. Self-assembly of surface functionalized amphiphilic carbon dots: tuning in morphological manifestations. SOFT MATTER 2019; 15:2863-2875. [PMID: 30840017 DOI: 10.1039/c9sm00051h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Despite the continuous surge of interest in supramolecular chemistry, the design and synthesis of building blocks to develop diverse examples of self-assemblies is still challenging. During the past decades, formation of self-assemblies such as micelles, vesicles, and gels with a fibril network using amphiphiles has been investigated at length. Considering the increasing applications of these self-aggregates across the scientific domain, it is crucial to adopt an alternative strategy for the preparation of self-aggregates using a new building block that has been applied in diverse domains. With this aim, surface functionalized carbon dots (CDs) with varying aliphatic/aromatic (cholesteryl, palmitoyl, naphthyl) substitutions linked with spacers such as ethylenediamine, p-phenylenediamine, 2,2'-(ethylenedioxy)bis(ethylamine) were developed. The surface passivated CDs formed self-assemblies in dimethylsulfoxide-water (DMSO-H2O, 2 : 1, v/v). The roles of surface functionalities and spacer units in the formation of self-assemblies using the synthesized CDs were investigated by microscopic and spectroscopic studies. Progressive morphological transition was found from vesicle-to-fiber in DMSO-H2O (2 : 1, v/v) which was dependent on surface passivating substitutions of the CDs from cholesteryl to naphthyl to palmitoyl. Together with the exclusive formation of self-assemblies using amphiphilic CDs, the present study enabled the tuning of self-organization behaviour of the CD by alteration of its surface functionality.
Collapse
Affiliation(s)
- Saheli Sarkar
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| | - Soumik Dinda
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| | - Pritam Choudhury
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| |
Collapse
|
48
|
Dai Y, Su J, Wu K, Ma W, Wang B, Li M, Sun P, Shen Q, Wang Q, Fan Q. Multifunctional Thermosensitive Liposomes Based on Natural Phase-Change Material: Near-Infrared Light-Triggered Drug Release and Multimodal Imaging-Guided Cancer Combination Therapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:10540-10553. [PMID: 30807086 DOI: 10.1021/acsami.8b22748] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Multifunctional theranostic nanoplatforms (NPs) in response to environment stimulations for on-demand drug release are highly desirable. Herein, the near-infrared (NIR)-absorbing dye, indocyanine green (ICG), and the antitumor drug, doxorubicin (DOX), were efficiently coencapsulated into the thermosensitive liposomes based on natural phase-change material. Folate and conjugated gadolinium (Gd) chelate-modified liposome shells enhance active targeting and magnetic resonance performance of the NPs while maintaining the size of the NPs. The ICG/DOX-loaded and gadolinium chelate conjugated temperature-sensitive liposome nanoplatforms (ID@TSL-Gd NPs) exhibited NIR-triggered drug release and prominent chemo-, photothermal, and photodynamic therapy properties. With the coencapsulated ICG, DOX, and the conjugated gadolinium chelates, the ID@TSL-Gd NPs can be used for triple-modal imaging (fluorescence/photoacoustic/magnetic resonance imaging)-guided combination tumor therapy (chemotherapy, photothermotherapy, and photodynamic therapy). After tail vein injection, the ID@TSL-Gd NPs accumulated effectively in subcutaneous HeLa tumor of mice. The tumor was effectively suppressed by accurate imaging-guided NIR-triggered phototherapy and chemotherapy, and no tumor regression and side effects were observed. In summary, the prepared ID@TSL-Gd NPs achieved multimodal imaging-guided cancer combination therapy, providing a promising platform for improving diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Yeneng Dai
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Jinzhong Su
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Kun Wu
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Wenkang Ma
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Bing Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Meixing Li
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Pengfei Sun
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Qingming Shen
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Qi Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM) , Nanjing University of Posts & Telecommunications , Nanjing 210023 , China
| |
Collapse
|
49
|
van Ballegooie C, Man A, Win M, Yapp DT. Spatially Specific Liposomal Cancer Therapy Triggered by Clinical External Sources of Energy. Pharmaceutics 2019; 11:E125. [PMID: 30884786 PMCID: PMC6470770 DOI: 10.3390/pharmaceutics11030125] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 11/25/2022] Open
Abstract
This review explores the use of energy sources, including ultrasound, magnetic fields, and external beam radiation, to trigger the delivery of drugs from liposomes in a tumor in a spatially-specific manner. Each section explores the mechanism(s) of drug release that can be achieved using liposomes in conjunction with the external trigger. Subsequently, the treatment's formulation factors are discussed, highlighting the parameters of both the therapy and the medical device. Additionally, the pre-clinical and clinical trials of each triggered release method are explored. Lastly, the advantages and disadvantages, as well as the feasibility and future outlook of each triggered release method, are discussed.
Collapse
Affiliation(s)
- Courtney van Ballegooie
- Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada.
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Alice Man
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Mi Win
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada.
| | - Donald T Yapp
- Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
50
|
Ling L, Ismail M, Du Y, Yao C, Li X. Lipoic acid-derived cross-linked liposomes for reduction-responsive delivery of anticancer drug. Int J Pharm 2019; 560:246-260. [PMID: 30769133 DOI: 10.1016/j.ijpharm.2019.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/19/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022]
Abstract
Liposomes have emerged as a fascinating nanocarriers for the delivery of cancer therapeutics. However, their efficacy for cancer therapy is reduced partially because of the serum-instability and incomplete drug release. In this study, a novel disulfide cross-linked liposomes (CLs) assembled from dimeric lipoic acid-derived glycerophosphorylcholine (di-LA-PC) conjugate was developed. The conjugate was synthesized by a facial esterification of lipoic acid (LA) and glycerophosphorylcholine (GPC) and characterized by MS, 1H NMR and 13C NMR. Featuring the enhanced serum-stability and intracellular drug release determined by in vitro stability and GSH-responsive behavior, CLs prepared with dried thin film technique following 10 % dithiothreitol (DTT) cross-linking can attain effective delivery of anticancer candidates. Notably, CLs stably encapsulated doxorubicin (Dox) in their vesicular structures and showed a remarkable thiol-sensitive release of payload upon cellular uptake by cancer cells, compared to that of uncross-linked liposomes (uCLs) or Doxil-like liposome (DLLs). The cell viability and apoptosis of Dox-loaded CLs worked the pronounced cytotoxic effects to MCF-7 cells with an IC50 value of 10.8 μg Dox equiv./mL comparable to free Dox and 2.8-fold higher than DLLs. More importantly, it is demonstrated that the nanoscale characteristics of Dox-loaded CLs could prevent the proliferation of adriamycin-resistant MCF-7/ADR cell line, highlighting their potential in reversal of drug resistance. Furthermore, the preliminary in vivo test (n = 3) showed that disulfide cross-linked liposomal formulation of Dox (Dox-CLs) improved the therapeutic efficacy compared to free Dox and DLLs in a human breast carcinoma xenograft mouse model. Therefore, the current thiol-responsive cross-linked liposome may provide a robust drug delivery platform for cancer therapy.
Collapse
Affiliation(s)
- Longbing Ling
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Muhammad Ismail
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yawei Du
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Chen Yao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|