1
|
Cressey P, Wilson JC, Amrahli M, Thanou M. Liposomes for Magnetic Resonance Image-Guided Drug Delivery; Lipid Chain Length Affects Drug Release and MRI Relaxivity. Molecules 2025; 30:1729. [PMID: 40333620 PMCID: PMC12029585 DOI: 10.3390/molecules30081729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 05/09/2025] Open
Abstract
Image-guided drug delivery is a method for tracking drug carriers for activation in specific lesions in the body. Image guidance uses the labelling of the drug or carrier and a clinically approved imaging modality. MRI (magnetic resonance image)-guided drug delivery has been considered for focused ultrasound tumour-targeted drug release. Liposomes are labelled for MRI tracking and the confirmation of drug delivery. In this study, we prepared two lipids conjugated to Gd-DOTA that confer MR imaging properties. Two lipid conjugates to DOTA, a C18 (LCA-1) and a C16 (LCA-2), were synthesised. The lipids were combined at different ratios within the lipid mix, and we investigated their effects on the liposome's Tm using DSC (differential scanning calorimetry) and on relaxivity using NMR. The results show that when different combinations of LCA-1 and LCA-2 were introduced into the liposomes, their ratio affected both thermal drug release and relaxivity. As these lipids are part of the liposomal membrane, they confer tracking ability, and their effect on relaxivity due to thermal release could enable the confirmation of liposomal drug release using MRI at clinically relevant magnetic field strengths.
Collapse
Affiliation(s)
| | | | | | - Maya Thanou
- School of Cancer and Pharmaceutical Sciences, Institute of Pharmaceutical Science, King’s College London, London SE1 9NH, UK; (P.C.); (J.C.W.); (M.A.)
| |
Collapse
|
2
|
Bai Z, Li Z, Shao Y. Subcellular Cavitation Bubbles Induce Cellular Mechanolysis and Collective Wound Healing in Ultrasound-Inflicted Cell Ablation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410760. [PMID: 39887946 PMCID: PMC11923933 DOI: 10.1002/advs.202410760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/21/2024] [Indexed: 02/01/2025]
Abstract
Focused ultrasound (FUS) has been widely adopted in medical and life science researches. Although various physical and biological effects of FUS have been well-documented, there is still a lack of understanding and direct evidence on the biological mechanism of therapeutic cell ablation caused by high-intensity ultrasound (HIFU) and the subsequent wound healing responses. This study develops an enclosed cell culture device that synergistically combines non-invasive FUS stimulation and real-time, on-the-fly live-cell imaging, providing an in vitro platform to explore short and long-term biological effects of ultrasound. The process, mechanism, and wound healing response of cell ablation induced by HIFU are elucidated, revealing a unique mechanism, termed ultrasound-inflicted cellular mechanolysis, that is mediated by growing subcellular cavitation air bubbles under confined contact with cells. This provides a previously unappreciated mechanism for understanding the biomechanical principles of ultrasound-based ablative therapy. A post-ablation phantom layer is also revealed that serves as a guiding cue for collective cell migration during wound healing, thereby providing a biomimetic model for studying wound healing after HIFU-inflicted damage. Together, this study provides theoretical and technological basis for advancing the understanding of the biological effects of ultrasound-based ablative therapy and inspiring clinically relevant applications in the future.
Collapse
Affiliation(s)
- Ziyue Bai
- Institute of Biomechanics and Medical EngineeringApplied Mechanics LaboratoryDepartment of Engineering MechanicsSchool of Aerospace EngineeringTsinghua UniversityBeijing100084China
| | - Zaimeng Li
- Institute of Fluid MechanicsDepartment of Engineering MechanicsSchool of Aerospace EngineeringTsinghua UniversityBeijing100084China
| | - Yue Shao
- Institute of Biomechanics and Medical EngineeringApplied Mechanics LaboratoryDepartment of Engineering MechanicsSchool of Aerospace EngineeringTsinghua UniversityBeijing100084China
| |
Collapse
|
3
|
Liao Z, Liu T, Yao Z, Hu T, Ji X, Yao B. Harnessing stimuli-responsive biomaterials for advanced biomedical applications. EXPLORATION (BEIJING, CHINA) 2025; 5:20230133. [PMID: 40040822 PMCID: PMC11875454 DOI: 10.1002/exp.20230133] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/18/2024] [Indexed: 03/06/2025]
Abstract
Cell behavior is intricately intertwined with the in vivo microenvironment and endogenous pathways. The ability to guide cellular behavior toward specific goals can be achieved by external stimuli, notably electricity, light, ultrasound, and magnetism, simultaneously harnessed through biomaterial-mediated responses. These external triggers become focal points within the body due to interactions with biomaterials, facilitating a range of cellular pathways: electrical signal transmission, biochemical cues, drug release, cell loading, and modulation of mechanical stress. Stimulus-responsive biomaterials hold immense potential in biomedical research, establishing themselves as a pivotal focal point in interdisciplinary pursuits. This comprehensive review systematically elucidates prevalent physical stimuli and their corresponding biomaterial response mechanisms. Moreover, it delves deeply into the application of biomaterials within the domain of biomedicine. A balanced assessment of distinct physical stimulation techniques is provided, along with a discussion of their merits and limitations. The review aims to shed light on the future trajectory of physical stimulus-responsive biomaterials in disease treatment and outline their application prospects and potential for future development. This review is poised to spark novel concepts for advancing intelligent, stimulus-responsive biomaterials.
Collapse
Affiliation(s)
- Ziming Liao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjinP. R. China
| | - Tingting Liu
- Division of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolCambridgeMassachusettsUSA
- Research Center for Nano‐Biomaterials and Regenerative MedicineDepartment of Biomedical EngineeringCollege of Biomedical EngineeringTaiyuan University of TechnologyTaiyuanShanxiP. R. China
- Department of Laboratory DiagnosisThe 971th HospitalQingdaoP. R. China
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research DepartmentPLA General Hospital and PLA Medical CollegeBeijingP. R. China
| | - Zhimin Yao
- Sichuan Preschool Educators' CollegeMianyangP. R. China
| | - Tian Hu
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular Medicine, University of OxfordJohn Radcliffe HospitalOxfordUK
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjinP. R. China
| | - Bin Yao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjinP. R. China
| |
Collapse
|
4
|
Taheri Z, Mozafari N, Moradian G, Lovison D, Dehshahri A, De Marco R. Integrin-Specific Stimuli-Responsive Nanomaterials for Cancer Theranostics. Pharmaceutics 2024; 16:1441. [PMID: 39598564 PMCID: PMC11597626 DOI: 10.3390/pharmaceutics16111441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/01/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Cancer is one of the leading causes of death worldwide. The tumor microenvironment makes the tumor difficult to treat, favoring drug resistance and the formation of metastases, resulting in death. Methods: Stimuli-responsive nanoparticles have shown great capacity to be used as a powerful strategy for cancer treatment, diagnostic, as well as theranostic. Nanocarriers are not only able to respond to internal stimuli such as oxidative stress, weakly acidic pH, high temperature, and the high expression of particular enzymes, but also to external stimuli such as light and paramagnetic characteristics to be exploited. Results: In this work, stimulus-responsive nanocarriers functionalized with arginine-glycine-aspartic acid (Arg-Gly-Asp) sequence as well as mimetic sequences with the capability to recognize integrin receptors are analyzed. Conclusions: This review highlights the progress that has been made in the development of new nanocarriers, capable of responding to endogenous and exogenous stimuli essential to combat cancer.
Collapse
Affiliation(s)
- Zahra Taheri
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-17336, Iran; (Z.T.); (N.M.)
- Student Research Committee, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-17336, Iran;
| | - Negin Mozafari
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-17336, Iran; (Z.T.); (N.M.)
| | - Ghazal Moradian
- Student Research Committee, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-17336, Iran;
| | - Denise Lovison
- Department of Agricultural, Food, Environmental and Animal Sciences (Di4A), University of Udine, 33100 Udine, Italy;
| | - Ali Dehshahri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-17336, Iran
| | - Rossella De Marco
- Department of Agricultural, Food, Environmental and Animal Sciences (Di4A), University of Udine, 33100 Udine, Italy;
| |
Collapse
|
5
|
Yan S, Zheng D. A Deep Neural Network for Predicting Synergistic Drug Combinations on Cancer. Interdiscip Sci 2024; 16:218-230. [PMID: 38183569 DOI: 10.1007/s12539-023-00596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/19/2023] [Accepted: 11/21/2023] [Indexed: 01/08/2024]
Abstract
The exploration of drug combinations presents an opportunity to amplify therapeutic effectiveness while alleviating undesirable side effects. Nevertheless, the extensive array of potential combinations poses challenges in terms of cost and time constraints for experimental screening. Thus, it is crucial to narrow down the search space. Deep learning approaches have gained widespread popularity in predicting synergistic drug combinations tailored for specific cell lines in vitro settings. In the present study, we introduce a novel method termed GTextSyn, which utilizes the integration of gene expression data and chemical structure information for the prediction of synergistic effects in drug combinations. GTextSyn employs a sentence classification model within the domain of Natural Language Processing (NLP), wherein drugs and cell lines are regarded as entities possessing biochemical relevance. Meanwhile, combinations of drug pairs and cell lines are construed as sentences with biochemical relational significance. To assess the efficacy of GTextSyn, we conduct a comparative analysis with alternative deep learning approaches using a standard benchmark dataset. The results from a five-fold cross-validation demonstrate a 49.5% reduction in Mean Square Error (MSE) achieved by GTextSyn, surpassing the performance of the next best method in the regression task. Furthermore, we conduct a comprehensive literature survey on the predicted novel drug combinations and find substantial support from prior experimental studies for many of the combinations identified by GTextSyn.
Collapse
Affiliation(s)
- Shiyu Yan
- School of Computer, University of South China, West Changsheng Road, Hengyang, 421001, Hunan, China.
| | - Ding Zheng
- School of Computer, University of South China, West Changsheng Road, Hengyang, 421001, Hunan, China
| |
Collapse
|
6
|
Gawne PJ, Ferreira M, Papaluca M, Grimm J, Decuzzi P. New Opportunities and Old Challenges in the Clinical translation of Nanotheranostics. NATURE REVIEWS. MATERIALS 2023; 8:783-798. [PMID: 39022623 PMCID: PMC11251001 DOI: 10.1038/s41578-023-00581-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 07/20/2024]
Abstract
Nanoparticle-based systems imbued with both diagnostic and therapeutic functions, known as nanotheranostics, have enabled remarkable progress in guiding focal therapy, inducing active responses to endogenous and exogenous biophysical stimuli, and stratifying patients for optimal treatment. However, although in recent years more nanotechnological platforms and techniques have been implemented in the clinic, several important challenges remain that are specific to nanotheranostics. In this Review, we first discuss some of the many ways of 'constructing' nanotheranostics, focusing on the different imaging modalities and therapeutic strategies. We then outline nanotheranostics that are currently used in humans at different stages of clinical development, identifying specific advantages and opportunities. Finally, we define critical steps along the winding road of preclinical and clinical development and suggest actions to overcome technical, manufacturing, regulatory and economical challenges for the safe and effective clinical translation of nanotheranostics.
Collapse
Affiliation(s)
- Peter J. Gawne
- UCL Cancer Institute, University College London, London, UK
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary, University of London, London, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Miguel Ferreira
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Marisa Papaluca
- School of Public Health, Imperial College of London, South Kensington CampusLondon, UK
| | - Jan Grimm
- Molecular Pharmacology Program and Department of Radiology, Memorial Sloan-Kettering Cancer, Center, New York, NY, USA
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via, Morego 30, 16163, Genoa, IT
| |
Collapse
|
7
|
Kommineni N, Chaudhari R, Conde J, Tamburaci S, Cecen B, Chandra P, Prasad R. Engineered Liposomes in Interventional Theranostics of Solid Tumors. ACS Biomater Sci Eng 2023; 9:4527-4557. [PMID: 37450683 DOI: 10.1021/acsbiomaterials.3c00510] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Engineered liposomal nanoparticles have unique characteristics as cargo carriers in cancer care and therapeutics. Liposomal theranostics have shown significant progress in preclinical and clinical cancer models in the past few years. Liposomal hybrid systems have not only been approved by the FDA but have also reached the market level. Nanosized liposomes are clinically proven systems for delivering multiple therapeutic as well as imaging agents to the target sites in (i) cancer theranostics of solid tumors, (ii) image-guided therapeutics, and (iii) combination therapeutic applications. The choice of diagnostics and therapeutics can intervene in the theranostics property of the engineered system. However, integrating imaging and therapeutics probes within lipid self-assembly "liposome" may compromise their overall theranostics performance. On the other hand, liposomal systems suffer from their fragile nature, site-selective tumor targeting, specific biodistribution and premature leakage of loaded cargo molecules before reaching the target site. Various engineering approaches, viz., grafting, conjugation, encapsulations, etc., have been investigated to overcome the aforementioned issues. It has been studied that surface-engineered liposomes demonstrate better tumor selectivity and improved therapeutic activity and retention in cells/or solid tumors. It should be noted that several other parameters like reproducibility, stability, smooth circulation, toxicity of vital organs, patient compliance, etc. must be addressed before using liposomal theranostics agents in solid tumors or clinical models. Herein, we have reviewed the importance and challenges of liposomal medicines in targeted cancer theranostics with their preclinical and clinical progress and a translational overview.
Collapse
Affiliation(s)
- Nagavendra Kommineni
- Center for Biomedical Research, Population Council, New York, New York 10065, United States
| | - Ruchita Chaudhari
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - João Conde
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa; Lisboa 1169-056, Portugal
| | - Sedef Tamburaci
- Department of Chemical Engineering, Izmir Institute of Technology, Gulbahce Campus, Izmir 35430, Turkey
| | - Berivan Cecen
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rajendra Prasad
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| |
Collapse
|
8
|
Zhang P, Ye G, Xie G, Lv J, Zeng X, Jiang W. Research progress of nanomaterial drug delivery in tumor targeted therapy. Front Bioeng Biotechnol 2023; 11:1240529. [PMID: 37555076 PMCID: PMC10405625 DOI: 10.3389/fbioe.2023.1240529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023] Open
Abstract
Cancer is one of the most lethal diseases in human society, and its incidence is gradually increasing. However, the current tumor treatment often meets the problem of poor efficacy and big side effects. The unique physical and chemical properties of nanomaterials can target the delivery of drugs to tumors, which can improve the therapeutic effect while reducing the damage of drugs to normal cells. This makes nanomaterials become a hot topic in the field of biomedicine. This review summarizes the recent progress of nanomaterials in tumor targeted therapy.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, China
| | - Guihua Ye
- Shanghai Ninth People’s Hospital Hainan Branch, Hainan Western Central Hospital, Danzhou, China
| | - Guofeng Xie
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jie Lv
- School of Computer Science and Engineering, Yulin Normal University, Yulin, China
| | - Xianhai Zeng
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, China
| | - Wei Jiang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Abuhelal S, Centelles MN, Wright M, Mason AJ, Thanou M. Development of Cationic Lipid LAH4-L1 siRNA Complexes for Focused Ultrasound Enhanced Tumor Uptake. Mol Pharm 2023; 20:2341-2351. [PMID: 36989421 PMCID: PMC10155207 DOI: 10.1021/acs.molpharmaceut.2c00909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
RNAi has considerable potential as a cancer therapeutic approach, but effective and efficient delivery of short interfering RNA (siRNA) to tumors remains a major hurdle. It remains a challenge to prepare a functional siRNA complex, target enough dose to the tumor, and stimulate its internalization into tumor cells and its release to the cytoplasm. Here, we show how these key barriers to siRNA delivery can be overcome with a complex─comprising siRNA, cationic lipids, and pH-responsive peptides─that is suited to tumor uptake enhancement via focused ultrasound (FUS). The complex provides effective nucleic acid encapsulation, nuclease protection, and endosomal escape such that gene silencing in cells is substantially more effective than that obtained with either equivalent lipoplexes or commercial reagents. In mice bearing MDA-MB-231 breast cancer xenografts, both lipid and ternary, lipid:peptide:siRNA complexes, prepared with near-infrared fluorescently labeled siRNA, accumulate in tumors following FUS treatments. Therefore, combining a well-designed lipid:peptide:siRNA complex with FUS tumor treatments is a promising route to achieve robust in vivo gene delivery.
Collapse
Affiliation(s)
- Shahd Abuhelal
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Miguel N Centelles
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Michael Wright
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - A James Mason
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| | - Maya Thanou
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, U.K
| |
Collapse
|
10
|
Aneja P, Negi P, Aneja S, Garad SR, Kumar S. Formulation optimization of pH-sensitive liposomes based drug delivery of Carboplatin and anti-proliferative evaluation against A549 (human lung carcinoma) cell lines. MAIN GROUP CHEMISTRY 2023. [DOI: 10.3233/mgc-220078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
The developments of pH–sensitive liposomes which are stable at physiological pH i.e. (6.8–7.4) have not explored much up until now. These lipid vesicles will go through destabilization and attain fusogenic properties in acidic conditions leading to liberation of aqueous contents. Carboplatin, included in the family of alkylating agent was found to exhibit adverse effects like myelo suppression, ion thrombocytopenia and leucopenia. Therefore, in order to circumvent these effects, carboplatin pH-sensitive liposomes for specific delivery is the ideal criteria and it poses a great challenge since the water-soluble drugs exhibited very low entrapment efficiency. The essential portion of study was evaluated using the Design Expert software 8. The pH-sensitive liposomes were optimized using Central composite design and one factor Response surface model design method and were prepared by film hydration method. Two formulation variables like drug: lipid ratio (X1) and volume of hydration media (X2) used to vary at three different levels and the other three variables viz. temperature, speed of rotation and vacuum applied were kept constant. The Response surface and contour plots were figured to elicit the effects of interaction of variables on the overall entrapment efficiency. pH-sensitive liposomes of carboplatin have been regarded as a promising delivery systematic approach in order to target tumor tissue as evaluated by the pre-clinical studies in both in vitro and ex-vivo conditions.
Collapse
Affiliation(s)
- Preeti Aneja
- Department of Pharmaceutics, Abhilashi College of Pharmacy, Nerchowk, Mandi, Himachal Pradesh, India
| | - Prabhat Negi
- Department of Pharmaceutics, Abhilashi College of Pharmacy, Nerchowk, Mandi, Himachal Pradesh, India
| | - Shivali Aneja
- Department of Microbiology, Shoolini University Solan, Himachal Pradesh, India
| | | | - Sunil Kumar
- Department of Chemistry, J.C. Bose University of Science and Technology, YMCA, Faridabad, Haryana, India
| |
Collapse
|
11
|
Hughes KA, Misra B, Maghareh M, Bobbala S. Use of stimulatory responsive soft nanoparticles for intracellular drug delivery. NANO RESEARCH 2023; 16:6974-6990. [PMID: 36685637 PMCID: PMC9840428 DOI: 10.1007/s12274-022-5267-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 05/24/2023]
Abstract
Drug delivery has made tremendous advances in the last decade. Targeted therapies are increasingly common, with intracellular delivery highly impactful and sought after. Intracellular drug delivery systems have limitations due to imprecise and non-targeted release profiles. One way this can be addressed is through using stimuli-responsive soft nanoparticles, which contain materials with an organic backbone such as lipids and polymers. The choice of biomaterial is essential for soft nanoparticles to be responsive to internal or external stimuli. The nanoparticle must retain its integrity and payload in non-targeted physiological conditions while responding to particular intracellular environments where payload release is desired. Multiple internal and external factors could stimulate the intracellular release of drugs from nanoparticles. Internal stimuli include pH, oxidation, and enzymes, while external stimuli include ultrasound, light, electricity, and magnetic fields. Stimulatory responsive soft nanoparticulate systems specifically utilized to modulate intracellular delivery of drugs are explored in this review.
Collapse
Affiliation(s)
- Krystal A. Hughes
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Maryam Maghareh
- Department of Clinical Pharmacy, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| |
Collapse
|
12
|
Image-guided drug delivery in nanosystem-based cancer therapies. Adv Drug Deliv Rev 2023; 192:114621. [PMID: 36402247 DOI: 10.1016/j.addr.2022.114621] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/18/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
The past decades have shown significant advancements in the development of solid tumor treatment. For instance, implementation of nanosystems for drug delivery has led to a reduction in side effects and improved delivery to the tumor region. However, clinical translation has faced challenges, as tumor drug levels are still considered to be inadequate. Interdisciplinary research has resulted in the development of more advanced drug delivery systems. These are coined "smart" due to the ability to be followed and actively manipulated in order to have better control over local drug release. Therefore, image-guided drug delivery can be a powerful strategy to improve drug activity at the target site. Being able to visualize the inflow of the administered smart nanosystem within the tumor gives the potential to determine the right moment to apply the facilitator to initiate drug release. Here we provide an overview of available nanosystems, imaging moieties, and imaging techniques. We discuss preclinical application of these smart drug delivery systems, the strength of image-guided drug delivery, and the future of personalized treatment.
Collapse
|
13
|
Pallares RM, Mottaghy FM, Schulz V, Kiessling F, Lammers T. Nanoparticle Diagnostics and Theranostics in the Clinic. J Nucl Med 2022; 63:1802-1808. [PMID: 36302654 PMCID: PMC9730918 DOI: 10.2967/jnumed.122.263895] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/19/2022] [Indexed: 01/11/2023] Open
Abstract
Nanoparticles possess unique features that may be useful for disease diagnosis and therapy. Preclinically, many different nanodiagnostics have been explored, but only a few have made it to the market. We here provide an overview of nanoparticle-based imaging agents currently used and evaluated in the clinic and discuss preclinical progress and translational avenues for the use of nanoparticles for diagnostic and theranostic applications.
Collapse
Affiliation(s)
- Roger M. Pallares
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Felix M. Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany;,Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands; and
| | - Volkmar Schulz
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany;,Physics Institute III B, RWTH Aachen University, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
14
|
Ashrafizadeh M, Delfi M, Zarrabi A, Bigham A, Sharifi E, Rabiee N, Paiva-Santos AC, Kumar AP, Tan SC, Hushmandi K, Ren J, Zare EN, Makvandi P. Stimuli-responsive liposomal nanoformulations in cancer therapy: Pre-clinical & clinical approaches. J Control Release 2022; 351:50-80. [PMID: 35934254 DOI: 10.1016/j.jconrel.2022.08.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
The site-specific delivery of antitumor agents is of importance for providing effective cancer suppression. Poor bioavailability of anticancer compounds and the presence of biological barriers prevent their accumulation in tumor sites. These obstacles can be overcome using liposomal nanostructures. The challenges in cancer chemotherapy and stimuli-responsive nanocarriers are first described in the current review. Then, stimuli-responsive liposomes including pH-, redox-, enzyme-, light-, thermo- and magneto-sensitive nanoparticles are discussed and their potential for delivery of anticancer drugs is emphasized. The pH- or redox-sensitive liposomes are based on internal stimulus and release drug in response to a mildly acidic pH and GSH, respectively. The pH-sensitive liposomes can mediate endosomal escape via proton sponge. The multifunctional liposomes responsive to both redox and pH have more capacity in drug release at tumor site compared to pH- or redox-sensitive alone. The magnetic field and NIR irradiation can be exploited for external stimulation of liposomes. The light-responsive liposomes release drugs when they are exposed to irradiation; thermosensitive-liposomes release drugs at a temperature of >40 °C when there is hyperthermia; magneto-responsive liposomes release drugs in presence of magnetic field. These smart nanoliposomes also mediate co-delivery of drugs and genes in synergistic cancer therapy. Due to lack of long-term toxicity of liposomes, they can be utilized in near future for treatment of cancer patients.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey.
| | - Masoud Delfi
- Department of Chemical Sciences, University of Naples "Federico II", Complesso Universitario Monte S. Angelo, Via Cintia, Naples 80126, Italy
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials-National Research Council (IPCB-CNR), Viale J.F. Kennedy 54-Mostra d'Oltremare pad. 20, 80125 Naples, Italy
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Navid Rabiee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea; School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology and zoonosis, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | | | - Pooyan Makvandi
- School of Chemistry, Damghan University, Damghan 36716-41167, Iran; Istituto Italiano di Tecnologia, Center for Materials Interfaces, viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy.
| |
Collapse
|
15
|
Singh E, Banerjee R. In vivo efficacy & phantom imaging connote the theranostic potential of a drug-loaded lipid nanobubble. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
16
|
Wang Z, Li J, Lin G, He Z, Wang Y. Metal complex-based liposomes: Applications and prospects in cancer diagnostics and therapeutics. J Control Release 2022; 348:1066-1088. [PMID: 35718211 DOI: 10.1016/j.jconrel.2022.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 06/09/2022] [Indexed: 12/17/2022]
Abstract
Metal complexes are of increasing interest as pharmaceutical agents in cancer diagnostics and therapeutics, while some of them suffer from issues such as limited water solubility and severe systemic toxicity. These drawbacks severely hampered their efficacy and clinical applications. Liposomes hold promise as delivery vehicles for constructing metal complex-based liposomes to maximize the therapeutic efficacy and minimize the side effects of metal complexes. This review provides an overview on the latest advances of metal complex-based liposomal delivery systems. First, the development of metal complex-mediated liposomal encapsulation is briefly introduced. Next, applications of metal complex-based liposomes in a variety of fields are overviewed, where drug delivery, cancer imaging (single photon emission computed tomography (SPECT), positron emission tomography (PET), and magnetic resonance imaging (MRI)), and cancer therapy (chemotherapy, phototherapy, and radiotherapy) were involved. Moreover, the potential toxicity, action of toxic mechanisms, immunological effects of metal complexes as well as the advantages of metal complex-liposomes in this content are also discussed. In the end, the future expectations and challenges of metal complex-based liposomes in clinical cancer therapy are tentatively proposed.
Collapse
Affiliation(s)
- Zhaomeng Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Guimei Lin
- School of Pharmacy, Shandong University, Jinan 250000, PR China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
17
|
Chaudhry M, Lyon P, Coussios C, Carlisle R. Thermosensitive liposomes: A promising step towards locsalised chemotherapy. Expert Opin Drug Deliv 2022; 19:899-912. [PMID: 35830722 DOI: 10.1080/17425247.2022.2099834] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Many small molecules and biologic therapeutics have been developed for solid tumor therapy. However, the unique physiology of tumors makes the actual delivery of these drugs into the tumor mass inefficient. Such delivery requires transport from blood vessels, across the vasculature and into and through interstitial space within a tumor. This transportation is dependent on the physiochemical properties of the therapeutic agent and the biological properties of the tumour. It was hoped the application of nanoscale drug carrier systems would solve this problem. However, issues with poor tumor accumulation and limited drug release have impeded clinical impact. In response, these carrier systems have been redesigned to be paired with targetable external mechanical stimuli which can trigger much enhanced drug release and deposition. AREAS COVERED The pre-clinical and clinical progress of thermolabile drug carrier systems and the modalities used to trigger the release of their cargo, is assessed. EXPERT OPINION Combined application of mild hyperthermia and heat-responsive liposomal drug carriers has great potential utility. Clinical trials continue to progress this approach and serve to refine the technologies, dosing regimens and exposure parameters that will provide optimal patient benefit.
Collapse
Affiliation(s)
| | - Paul Lyon
- Nuffield Dept of Surgical Sciences, University of Oxford, Oxford, UK.,Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, Engineering Science, University of Oxford, Oxford, UK
| | - Robert Carlisle
- Institute of Biomedical Engineering, Engineering Science, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Lu S, Zhao P, Deng Y, Liu Y. Mechanistic Insights and Therapeutic Delivery through Micro/Nanobubble-Assisted Ultrasound. Pharmaceutics 2022; 14:pharmaceutics14030480. [PMID: 35335857 PMCID: PMC8954263 DOI: 10.3390/pharmaceutics14030480] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/12/2022] [Accepted: 02/19/2022] [Indexed: 02/05/2023] Open
Abstract
Ultrasound with low frequency (20–100 kHz) assisted drug delivery has been widely investigated as a non-invasive method to enhance the permeability and retention effect of drugs. The functional micro/nanobubble loaded with drugs could provide an unprecedented opportunity for targeted delivery. Then, ultrasound with higher intensity would locally burst bubbles and release agents, thus avoiding side effects associated with systemic administration. Furthermore, ultrasound-mediated destruction of micro/nanobubbles can effectively increase the permeability of vascular membranes and cell membranes, thereby not only increasing the distribution concentration of drugs in the interstitial space of target tissues but also promoting the penetration of drugs through cell membranes into the cytoplasm. These advancements have transformed ultrasound from a purely diagnostic utility into a promising theragnostic tool. In this review, we first discuss the structure and generation of micro/nanobubbles. Second, ultrasound parameters and mechanisms of therapeutic delivery are discussed. Third, potential biomedical applications of micro/nanobubble-assisted ultrasound are summarized. Finally, we discuss the challenges and future directions of ultrasound combined with micro/nanobubbles.
Collapse
|
19
|
Shariatinia Z. Big family of nano- and microscale drug delivery systems ranging from inorganic materials to polymeric and stimuli-responsive carriers as well as drug-conjugates. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
20
|
Dual Targeting of Cancer Cells and MMPs with Self-Assembly Hybrid Nanoparticles for Combination Therapy in Combating Cancer. Pharmaceutics 2021; 13:pharmaceutics13121990. [PMID: 34959271 PMCID: PMC8707712 DOI: 10.3390/pharmaceutics13121990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/05/2021] [Accepted: 11/19/2021] [Indexed: 01/21/2023] Open
Abstract
The co-delivery of chemotherapeutic agents and immune modulators to their targets remains to be a great challenge for nanocarriers. Here, we developed a hybrid thermosensitive nanoparticle (TMNP) which could co-deliver paclitaxel-loaded transferrin (PTX@TF) and marimastat-loaded thermosensitive liposomes (MMST/LTSLs) for the dual targeting of cancer cells and the microenvironment. TMNPs could rapidly release the two payloads triggered by the hyperthermia treatment at the site of tumor. The released PTX@TF entered cancer cells via transferrin-receptor-mediated endocytosis and inhibited the survival of tumor cells. MMST was intelligently employed as an immunomodulator to improve immunotherapy by inhibiting matrix metalloproteinases to reduce chemokine degradation and recruit T cells. The TMNPs promoted the tumor infiltration of CD3+ T cells by 2-fold, including memory/effector CD8+ T cells (4.2-fold) and CD4+ (1.7-fold), but not regulatory T cells. Our in vivo anti-tumor experiment suggested that TMNPs possessed the highest tumor growth inhibitory rate (80.86%) compared with the control group. We demonstrated that the nanoplatform could effectively inhibit the growth of tumors and enhance T cell recruitment through the co-delivery of paclitaxel and marimastat, which could be a promising strategy for the combination of chemotherapy and immunotherapy for cancer treatment.
Collapse
|
21
|
Priester MI, Curto S, van Rhoon GC, ten Hagen TLM. External Basic Hyperthermia Devices for Preclinical Studies in Small Animals. Cancers (Basel) 2021; 13:cancers13184628. [PMID: 34572855 PMCID: PMC8470307 DOI: 10.3390/cancers13184628] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The application of mild hyperthermia can be beneficial for solid tumor treatment by induction of sublethal effects on a tissue- and cellular level. When designing a hyperthermia experiment, several factors should be taken into consideration. In this review, multiple elementary hyperthermia devices are described in detail to aid standardization of treatment design. Abstract Preclinical studies have shown that application of mild hyperthermia (40–43 °C) is a promising adjuvant to solid tumor treatment. To improve preclinical testing, enhance reproducibility, and allow comparison of the obtained results, it is crucial to have standardization of the available methods. Reproducibility of methods in and between research groups on the same techniques is crucial to have a better prediction of the clinical outcome and to improve new treatment strategies (for instance with heat-sensitive nanoparticles). Here we provide a preclinically oriented review on the use and applicability of basic hyperthermia systems available for solid tumor thermal treatment in small animals. The complexity of these techniques ranges from a simple, low-cost water bath approach, irradiation with light or lasers, to advanced ultrasound and capacitive heating devices.
Collapse
Affiliation(s)
- Marjolein I. Priester
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.C.); (G.C.v.R.)
| | - Sergio Curto
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.C.); (G.C.v.R.)
| | - Gerard C. van Rhoon
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (S.C.); (G.C.v.R.)
| | - Timo L. M. ten Hagen
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Correspondence:
| |
Collapse
|
22
|
Liu J, Zhong S, Zhang L, Yi M, Liu X, Bing T, Zhang N, Shangguan D. A 4-aminonaphthalimide-based fluorescent traceable prodrug with excellent photoinduced cytotoxicity. Chem Commun (Camb) 2021; 57:6558-6561. [PMID: 34113937 DOI: 10.1039/d1cc01847g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A blue light activated anti-cancer prodrug, NST, was designed based on a photoactive 4-aminonaphthalimide derivative and an anticancer drug, 10-hydroxycamptothecin. NST was hard to be taken up by living cells and showed negligible dark cytotoxicity. The irradiation caused photocleavage of NST and resulted in high cytotoxicity.
Collapse
Affiliation(s)
- Jing Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shilong Zhong
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Lingling Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Mengwen Yi
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Xiangjun Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tao Bing
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nan Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China. and University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dihua Shangguan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China. and University of Chinese Academy of Sciences, Beijing, 100049, China and School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310013, China
| |
Collapse
|
23
|
Akash SZ, Lucky FY, Hossain M, Bepari AK, Rahman GMS, Reza HM, Sharker SM. Remote Temperature-Responsive Parafilm Dermal Patch for On-Demand Topical Drug Delivery. MICROMACHINES 2021; 12:mi12080975. [PMID: 34442597 PMCID: PMC8400996 DOI: 10.3390/mi12080975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022]
Abstract
The development of externally controlled drug delivery systems that can rapidly trigger drug release is widely expected to change the landscape of future drug carriers. In this study, a drug delivery system was developed for on-demand therapeutic effects. The thermoresponsive paraffin film can be loaded on the basis of therapeutic need, including local anesthetic (lidocaine) or topical antibiotic (neomycin), controlled remotely by a portable mini-heater. The application of mild temperature (45 °C) to the drug-loaded paraffin film allowed a rapid stimulus response within a short time (5 min). This system exploits regular drug release and the rapid generation of mild heat to trigger a burst release of 80% within 6 h of any locally administered drug. The in vitro drug release studies and in vivo therapeutic activity were observed for local anesthesia and wound healing using a neomycin-loaded film. The studies demonstrated on-demand drug release with minimized inflammation and microbial infection. This temperature-responsive drug-loaded film can be triggered remotely to provide flexible control of dose magnitude and timing. Our preclinical studies on these remotely adjustable drug delivery systems can significantly improve patient compliance and medical practice.
Collapse
|
24
|
Tehrani Fateh S, Moradi L, Kohan E, Hamblin MR, Shiralizadeh Dezfuli A. Comprehensive review on ultrasound-responsive theranostic nanomaterials: mechanisms, structures and medical applications. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:808-862. [PMID: 34476167 PMCID: PMC8372309 DOI: 10.3762/bjnano.12.64] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/15/2021] [Indexed: 05/03/2023]
Abstract
The field of theranostics has been rapidly growing in recent years and nanotechnology has played a major role in this growth. Nanomaterials can be constructed to respond to a variety of different stimuli which can be internal (enzyme activity, redox potential, pH changes, temperature changes) or external (light, heat, magnetic fields, ultrasound). Theranostic nanomaterials can respond by producing an imaging signal and/or a therapeutic effect, which frequently involves cell death. Since ultrasound (US) is already well established as a clinical imaging modality, it is attractive to combine it with rationally designed nanoparticles for theranostics. The mechanisms of US interactions include cavitation microbubbles (MBs), acoustic droplet vaporization, acoustic radiation force, localized thermal effects, reactive oxygen species generation, sonoluminescence, and sonoporation. These effects can result in the release of encapsulated drugs or genes at the site of interest as well as cell death and considerable image enhancement. The present review discusses US-responsive theranostic nanomaterials under the following categories: MBs, micelles, liposomes (conventional and echogenic), niosomes, nanoemulsions, polymeric nanoparticles, chitosan nanocapsules, dendrimers, hydrogels, nanogels, gold nanoparticles, titania nanostructures, carbon nanostructures, mesoporous silica nanoparticles, fuel-free nano/micromotors.
Collapse
Affiliation(s)
- Sepand Tehrani Fateh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lida Moradi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Kohan
- Department of Science, University of Kurdistan, Kurdistan, Sanandaj, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | | |
Collapse
|
25
|
Awad N, Paul V, AlSawaftah NM, ter Haar G, Allen TM, Pitt WG, Husseini GA. Ultrasound-Responsive Nanocarriers in Cancer Treatment: A Review. ACS Pharmacol Transl Sci 2021; 4:589-612. [PMID: 33860189 PMCID: PMC8033618 DOI: 10.1021/acsptsci.0c00212] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Indexed: 12/13/2022]
Abstract
The safe and effective delivery of anticancer agents to diseased tissues is one of the significant challenges in cancer therapy. Conventional anticancer agents are generally cytotoxins with poor pharmacokinetics and bioavailability. Nanocarriers are nanosized particles designed for the selectivity of anticancer drugs and gene transport to tumors. They are small enough to extravasate into solid tumors, where they slowly release their therapeutic load by passive leakage or biodegradation. Using smart nanocarriers, the rate of release of the entrapped therapeutic(s) can be increased, and greater exposure of the tumor cells to the therapeutics can be achieved when the nanocarriers are exposed to certain internally (enzymes, pH, and temperature) or externally (light, magnetic field, and ultrasound) applied stimuli that trigger the release of their load in a safe and controlled manner, spatially and temporally. This review gives a comprehensive overview of recent research findings on the different types of stimuli-responsive nanocarriers and their application in cancer treatment with a particular focus on ultrasound.
Collapse
Affiliation(s)
- Nahid
S. Awad
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Vinod Paul
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Nour M. AlSawaftah
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Gail ter Haar
- Joint
Department of Physics, The Institute of
Cancer Research and The Royal Marsden NHS Foundation Trust, London SM2 5NG, U.K.
| | - Theresa M. Allen
- Department
of Pharmacology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - William G. Pitt
- Department
of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| | - Ghaleb A. Husseini
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
26
|
Song F, Gao H, Li D, Petrov AV, Petrov VV, Wen D, Sukhorukov GB. Low intensity focused ultrasound responsive microcapsules for non-ablative ultrafast intracellular release of small molecules. J Mater Chem B 2021; 9:2384-2393. [PMID: 33554993 DOI: 10.1039/d0tb02788j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Focused ultrasound (FU) is in demand for clinical cancer therapy, but the possible thermal injury to the normal peripheral tissues limits the usage of the ablative FU for tumors with a large size; therefore research efforts have been made to minimize the possible side effects induced by the FU treatment. Non-ablative focused ultrasound assisted chemotherapy could open a new avenue for the development of cancer therapy technology. Here, low intensity focused ultrasound (LIFU) for controlled quick intracellular release of small molecules (Mw ≤ 1000 Da) without acute cell damage is demonstrated. The release is achieved by a composite poly(allylamine hydrochloride) (PAH)/poly-(sodium 4-styrenesulfonate) (PSS)/SiO2 microcapsules which are highly sensitive to LIFU and can be effectively broken by weak cavitation effects. Most PAH/PSS/SiO2 capsules in B50 rat neuronal cells can be ruptured and release rhodamine B (Rh-B) into the cytosol within only 30 s of 0.75 W cm-2 LIFU treatment, as demonstrated by the CLSM results. While the same LIFU treatment shows no obvious damage to cells, as proved by the live/dead experiment, showing that 90% of cells remain alive.
Collapse
Affiliation(s)
- Fengyan Song
- School of Aeronautic Science and Engineering, Beihang University, Beijing, 100191, P. R. China.
| | - Hui Gao
- School of Aeronautic Science and Engineering, Beihang University, Beijing, 100191, P. R. China.
| | - Danyang Li
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK.
| | - Arseniy V Petrov
- Educational Research Institute of Nanostructures and Biosystems, Saratov State University, 83 Astrakhanskaya Street, Saratov, 410012, Russia
| | - Vladimir V Petrov
- Educational Research Institute of Nanostructures and Biosystems, Saratov State University, 83 Astrakhanskaya Street, Saratov, 410012, Russia
| | - Dongsheng Wen
- School of Aeronautic Science and Engineering, Beihang University, Beijing, 100191, P. R. China. and School of Chemical and Processing Engineering, University of Leeds, Leeds, LS2 9JT, UK
| | - Gleb B Sukhorukov
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK.
| |
Collapse
|
27
|
Cressey P, Amrahli M, So PW, Gedroyc W, Wright M, Thanou M. Image-guided thermosensitive liposomes for focused ultrasound enhanced co-delivery of carboplatin and SN-38 against triple negative breast cancer in mice. Biomaterials 2021; 271:120758. [PMID: 33774525 DOI: 10.1016/j.biomaterials.2021.120758] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 12/20/2022]
Abstract
Triggerable nanocarriers have the potential to significantly improve the therapeutic index of existing anticancer agents. They allow for highly localised delivery and release of therapeutic cargos, reducing off-target toxicity and increasing anti-tumour activity. Liposomes may be engineered to respond to an externally applied stimulus such as focused ultrasound (FUS). Here, we report the first co-delivery of SN-38 (irinotecan's super-active metabolite) and carboplatin, using an MRI-visible thermosensitive liposome (iTSL). MR contrast enhancement was achieved by the incorporation of a gadolinium lipid conjugate in the liposome bilayer along with a dye-labelled lipid for near infrared fluorescence bioimaging. The resulting iTSL were successfully loaded with SN-38 in the lipid bilayer and carboplatin in the aqueous core - allowing co-delivery of both. The iTSL demonstrated both thermosensitivity and MR-imageability. In addition, they showed effective local targeted co-delivery of carboplatin and SN-38 after triggered release with brief FUS treatments. A single dosage induced significant improvement of anti-tumour activity (over either the free drugs or the iTSL without FUS-activation) in triple negative breast cancer xenografts tumours in mice.
Collapse
Affiliation(s)
- Paul Cressey
- School of Cancer & Pharmaceutical Sciences, King's College London, UK
| | - Maral Amrahli
- School of Cancer & Pharmaceutical Sciences, King's College London, UK
| | - Po-Wah So
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Wladyslaw Gedroyc
- Radiology Department, Imperial College Healthcare NHS Trust, London, UK
| | - Michael Wright
- School of Cancer & Pharmaceutical Sciences, King's College London, UK
| | - Maya Thanou
- School of Cancer & Pharmaceutical Sciences, King's College London, UK.
| |
Collapse
|
28
|
Mai X, Chang Y, You Y, He L, Chen T. Designing intelligent nano-bomb with on-demand site-specific drug burst release to synergize with high-intensity focused ultrasound cancer ablation. J Control Release 2021; 331:270-281. [DOI: 10.1016/j.jconrel.2020.09.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/29/2022]
|
29
|
Nicolson F, Kircher MF. Theranostics: Agents for Diagnosis and Therapy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00040-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
30
|
Amrahli M, Centelles M, Cressey P, Prusevicius M, Gedroyc W, Xu XY, So PW, Wright M, Thanou M. MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice. Nanotheranostics 2021; 5:125-142. [PMID: 33457192 PMCID: PMC7806456 DOI: 10.7150/ntno.52168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/30/2020] [Indexed: 01/12/2023] Open
Abstract
Rationale: Image-guided, triggerable, drug delivery systems allow for precisely placed and highly localised anti-cancer treatment. They contain labels for spatial mapping and tissue uptake tracking, providing key location and timing information for the application of an external stimulus to trigger drug release. High Intensity Focused Ultrasound (HIFU or FUS) is a non-invasive approach for treating small tissue volumes and is particularly effective at inducing drug release from thermosensitive nanocarriers. Here, we present a novel MR-imageable thermosensitive liposome (iTSL) for drug delivery to triple-negative breast cancers (TNBC). Methods: A macrocyclic gadolinium-based Magnetic Resonance Imaging (MRI) contrast agent was covalently linked to a lipid. This was incorporated at 30 mol% into the lipid bilayer of a thermosensitive liposome that was also encapsulating doxorubicin. The resulting iTSL-DOX formulation was assessed for physical and chemical properties, storage stability, leakage of gadolinium or doxorubicin, and thermal- or FUS-induced drug release. Its effect on MRI relaxation time was tested in phantoms. Mice with tumours were used for studies to assess both tumour distribution and contrast enhancement over time. A lipid-conjugated near-infrared fluorescence (NIRF) probe was also included in the liposome to facilitate the real time monitoring of iTSL distribution and drug release in tumours by NIRF bioimaging. TNBC (MDA-MB-231) tumour-bearing mice were then used to demonstrate the efficacy at retarding tumour growth and increasing survival. Results: iTSL-DOX provided rapid FUS-induced drug release that was dependent on the acoustic power applied. It was otherwise found to be stable, with minimum leakage of drug and gadolinium into buffers or under challenging conditions. In contrast to the usually suggested longer FUS treatment we identified that brief (~3 min) FUS significantly enhanced iTSL-DOX uptake to a targeted tumour and triggered near-total release of encapsulated doxorubicin, causing significant growth inhibition in the TNBC mouse model. A distinct reduction in the tumours' average T1 relaxation times was attributed to the iTSL accumulation. Conclusions: We demonstrate that tracking iTSL in tumours using MRI assists the application of FUS for precise drug release and therapy.
Collapse
Affiliation(s)
- Maral Amrahli
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| | - Miguel Centelles
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| | - Paul Cressey
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| | | | | | - Xiao Yun Xu
- Department of Chemical Engineering, Imperial College London, U.K
| | - Po-Wah So
- Department of Neuroimaging, King's College London, U.K
| | - Michael Wright
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| | - Maya Thanou
- School of Cancer & Pharmaceutical Sciences, King's College London, U.K
| |
Collapse
|
31
|
Enhanced In Vitro Magnetic Cell Targeting of Doxorubicin-Loaded Magnetic Liposomes for Localized Cancer Therapy. NANOMATERIALS 2020; 10:nano10112104. [PMID: 33114052 PMCID: PMC7690690 DOI: 10.3390/nano10112104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022]
Abstract
The lack of efficient targeting strategies poses significant limitations on the effectiveness of chemotherapeutic treatments. This issue also affects drug-loaded nanocarriers, reducing nanoparticles cancer cell uptake. We report on the fabrication and in vitro characterization of doxorubicin-loaded magnetic liposomes for localized treatment of liver malignancies. Colloidal stability, superparamagnetic behavior and efficient drug loading of our formulation were demonstrated. The application of an external magnetic field guaranteed enhanced nanocarriers cell uptake under cell medium flow in correspondence of a specific area, as we reported through in vitro investigation. A numerical model was used to validate experimental data of magnetic targeting, proving the possibility of accurately describing the targeting strategy and predict liposomes accumulation under different environmental conditions. Finally, in vitro studies on HepG2 cancer cells confirmed the cytotoxicity of drug-loaded magnetic liposomes, with cell viability reduction of about 50% and 80% after 24 h and 72 h of incubation, respectively. Conversely, plain nanocarriers showed no anti-proliferative effects, confirming the formulation safety. Overall, these results demonstrated significant targeting efficiency and anticancer activity of our nanocarriers and superparamagnetic nanoparticles entrapment could envision the theranostic potential of the formulation. The proposed magnetic targeting study could represent a valid tool for pre-clinical investigation regarding the effectiveness of magnetic drug targeting.
Collapse
|
32
|
Duraisamy GS, Bhosale D, Lipenská I, Huvarova I, Růžek D, Windisch MP, Miller AD. Advanced Therapeutics, Vaccinations, and Precision Medicine in the Treatment and Management of Chronic Hepatitis B Viral Infections; Where Are We and Where Are We Going? Viruses 2020; 12:v12090998. [PMID: 32906840 PMCID: PMC7552065 DOI: 10.3390/v12090998] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
The management of chronic hepatitis B virus (CHB) infection is an area of massive unmet clinical need worldwide. In spite of the development of powerful nucleoside/nucleotide analogue (NUC) drugs, and the widespread use of immune stimulators such as interferon-alpha (IFNα) or PEGylated interferon-alpha (PEG-IFNα), substantial improvements in CHB standards of care are still required. We believe that the future for CHB treatment now rests with advanced therapeutics, vaccination, and precision medicine, if all are to bring under control this most resilient of virus infections. In spite of a plethora of active drug treatments, anti-viral vaccinations and diagnostic techniques, the management of CHB infection remains unresolved. The reason for this is the very complexity of the virus replication cycle itself, giving rise to multiple potential targets for therapeutic intervention some of which remain very intractable indeed. Our review is focused on discussing the potential impact that advanced therapeutics, vaccinations and precision medicine could have on the future management of CHB infection. We demonstrate that advanced therapeutic approaches for the treatment of CHB, in the form of gene and immune therapies, together with modern vaccination strategies, are now emerging rapidly to tackle the limitations of current therapeutic approaches to CHB treatment in clinic. In addition, precision medicine approaches are now gathering pace too, starting with personalized medicine. On the basis of this, we argue that the time has now come to accelerate the design and creation of precision therapeutic approaches (PTAs) for CHB treatment that are based on advanced diagnostic tools and nanomedicine, and which could maximize CHB disease detection, treatment, and monitoring in ways that could genuinely eliminate CHB infection altogether.
Collapse
Affiliation(s)
- Ganesh Selvaraj Duraisamy
- Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic; (G.S.D.); (D.B.); (I.L.); (I.H.); (D.R.)
| | - Dattatry Bhosale
- Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic; (G.S.D.); (D.B.); (I.L.); (I.H.); (D.R.)
| | - Ivana Lipenská
- Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic; (G.S.D.); (D.B.); (I.L.); (I.H.); (D.R.)
| | - Ivana Huvarova
- Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic; (G.S.D.); (D.B.); (I.L.); (I.H.); (D.R.)
| | - Daniel Růžek
- Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic; (G.S.D.); (D.B.); (I.L.); (I.H.); (D.R.)
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 České Budějovice, Czech Republic
| | - Marc P. Windisch
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, 696 Sampyeong-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Korea;
- Division of Bio-Medical Science and Technology, University of Science and Technology, Daejeon 305-350, Korea
| | - Andrew D. Miller
- Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic; (G.S.D.); (D.B.); (I.L.); (I.H.); (D.R.)
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemědělská 1, Černá Pole, CZ-61300 Brno, Czech Republic
- KP Therapeutics (Europe) s.r.o., Purkyňova 649/127, CZ-61200 Brno, Czech Republic
- Correspondence:
| |
Collapse
|
33
|
Sun T, Dasgupta A, Zhao Z, Nurunnabi M, Mitragotri S. Physical triggering strategies for drug delivery. Adv Drug Deliv Rev 2020; 158:36-62. [PMID: 32589905 DOI: 10.1016/j.addr.2020.06.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/06/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Physically triggered systems hold promise for improving drug delivery by enhancing the controllability of drug accumulation and release, lowering non-specific toxicity, and facilitating clinical translation. Several external physical stimuli including ultrasound, light, electric fields and magnetic fields have been used to control drug delivery and they share some common features such as spatial targeting, spatiotemporal control, and minimal invasiveness. At the same time, they possess several distinctive features in terms of interactions with biological entities and/or the extent of stimulus response. Here, we review the key advances of such systems with a focus on discussing their physical mechanisms, the design rationales, and translational challenges.
Collapse
Affiliation(s)
- Tao Sun
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anshuman Dasgupta
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Zongmin Zhao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, TX 79902, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Wang S, Mao J, Liu H, Huang S, Cai J, Gui W, Wu J, Xu J, Shen J, Wang Z. pH-Sensitive nanotheranostics for dual-modality imaging guided nanoenzyme catalysis therapy and phototherapy. J Mater Chem B 2020; 8:4859-4869. [PMID: 32100793 DOI: 10.1039/c9tb02731a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Triple-negative breast cancer shows resistance to conventional radiotherapies and chemotherapies, and few molecular targeted therapies are currently available for this malignancy in clinical settings. In this work, a theranostic nanosystem with a core-shell structure was synthesized through self-assembly of amphiphilic macromolecules, which can be used in effective catalysis therapy and phototherapy. Herein, we report that superparamagnetic iron oxide nanocrystals and IR780 dye were effectively loaded into the hydrophobic core of the nanosystem, which enabled dual-modality magnetic resonance imaging and near-infrared fluorescence imaging. Furthermore, on the hydrophilic crown, the neighboring carboxylic acid-based amide linkage showed charge conversion properties in the tumor acidic microenvironment and endowed the system with targeted diagnosis and delivery. Upon light excitation, this theranostic system exerted synergistic anti-cancer activity through the nano-enzyme catalysis effect enhanced phototherapy in a triple-negative 4T1 breast cancer model. Furthermore, there were no obvious side-effects. The results of our study demonstrate the great potential of this theranostic nanosystem in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Sheng Wang
- Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Jiaji Mao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Hong Liu
- Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Shihui Huang
- Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Jiali Cai
- Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Wentao Gui
- Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Jun Wu
- School of Medical Imaging, North Sichuan Medical College, Nanchong, 637000, China
| | - Junyao Xu
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Zhiyong Wang
- Center for Functional Biomaterials, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| |
Collapse
|
35
|
Meng Q, Cong H, Hu H, Xu FJ. Rational design and latest advances of codelivery systems for cancer therapy. Mater Today Bio 2020; 7:100056. [PMID: 32510051 PMCID: PMC7264083 DOI: 10.1016/j.mtbio.2020.100056] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/25/2020] [Accepted: 04/29/2020] [Indexed: 01/06/2023] Open
Abstract
Current treatments have limited effectiveness in treating tumors. The combination of multiple drugs or treatment strategies is widely studied to improve therapeutic effect and reduce adverse effects of cancer therapy. The codelivery system is the key to realize combined therapies. It is necessary to design and construct different codelivery systems in accordance with the variable structures and properties of cargoes and vectors. This review presented the typical design considerations about codelivery vectors for cancer therapy and described the current state of codelivery systems from two aspects: different types of vectors and collaborative treatment strategies. The commonly used loading methods of cargoes into the vectors, including physical and chemical processes, are discussed in detail. Finally, we outline the challenges and perspectives about the improvement of codelivery systems.
Collapse
Affiliation(s)
- Q.Y. Meng
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - H.L. Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - H. Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - F.-J. Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
36
|
Optimization of power used in liver cancer microwave therapy by injection of Magnetic Nanoparticles (MNPs). Comput Biol Med 2020; 120:103741. [DOI: 10.1016/j.compbiomed.2020.103741] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 02/08/2023]
|
37
|
Ahmad T, Sarwar R, Iqbal A, Bashir U, Farooq U, Halim SA, Khan A, Al-Harrasi A. Recent advances in combinatorial cancer therapy via multifunctionalized gold nanoparticles. Nanomedicine (Lond) 2020; 15:1221-1237. [DOI: 10.2217/nnm-2020-0051] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The diverse behavior of nanogold in the therapeutic field is related to its unique size and shape. Nanogold offers improvements in modern diagnostic and therapeutic implications, increases disease specificity and targeted drug delivery, and is relatively economical compared with other chemotherapeutic protocols. The diagnosis of cancer and photothermal therapy improve drastically with the implementation of nanotechnology. Different types of nanoparticles, that is, gold silica nanoshells, nanorods and nanospheres of diverse shapes and geometries, are used widely in the photothermal therapy of cancerous cells and nodules. Numerous reviews have been published on the therapeutic applications of gold nanoparticles, but studies on combinatorial applications of nanogold in cancer therapy are limited. This review focuses on the combinatorial cancer therapy using optical properties of nanogold with different shapes and geometries, and their therapeutic applications in cancer diagnosis, photothermal therapy, cancer imaging and targeted drug delivery.
Collapse
Affiliation(s)
- Touqeer Ahmad
- Natural & Medical Sciences Research Center, University of Nizwa, PO Box 33, Birkat Al Mauz, Nizwa, 616, Sultanate of Oman
| | - Rizwana Sarwar
- Department of Chemistry, COMSATS University Islamabad Abbottabad Campus, Abbottabad, Pakistan
| | - Ayesha Iqbal
- Division of Pharmacy Practice & Policy, School of pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Uzma Bashir
- Department of Chemistry, COMSATS University Islamabad Abbottabad Campus, Abbottabad, Pakistan
| | - Umar Farooq
- Department of Chemistry, COMSATS University Islamabad Abbottabad Campus, Abbottabad, Pakistan
| | - Sobia Ahsan Halim
- Natural & Medical Sciences Research Center, University of Nizwa, PO Box 33, Birkat Al Mauz, Nizwa, 616, Sultanate of Oman
| | - Ajmal Khan
- Natural & Medical Sciences Research Center, University of Nizwa, PO Box 33, Birkat Al Mauz, Nizwa, 616, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, PO Box 33, Birkat Al Mauz, Nizwa, 616, Sultanate of Oman
| |
Collapse
|
38
|
Liu J, Gao Y, Zhu Y, Zhu J, Wang C, Rui X, Yang K, Si D, Lin J, Yuan D, Wen H, Li W. Rhodium(III)-Catalyzed Oxidative Annulation of 4-Aminoquinolines and Acrylate through Two Consecutive C(sp 2)-H Activations. Org Lett 2020; 22:2657-2662. [PMID: 32186885 DOI: 10.1021/acs.orglett.0c00630] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The C-H annulation of the five-position of quinolines and acrylates to afford heterocycles is an active field of research in organic synthesis. Herein the annulation of 4-aminoquinolines with acrylates through two consecutive C-H activations catalyzed by Rh(III) is described. The reaction proceeds with high atom efficiency under mild reaction conditions, and this protocol will provide appealing strategies for the synthesis of fused quinoline heterocycles.
Collapse
Affiliation(s)
- Jian Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yi Gao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yehua Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Junru Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chao Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiyan Rui
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Kundi Yang
- Department of Chemistry and Biochemistry, Miami University, Oxford, Ohio 45056, United States
| | - Dongjuan Si
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiamin Lin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Dandan Yuan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hongmei Wen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
39
|
Rezaeian M, Sedaghatkish A, Soltani M. Numerical modeling of high-intensity focused ultrasound-mediated intraperitoneal delivery of thermosensitive liposomal doxorubicin for cancer chemotherapy. Drug Deliv 2020; 26:898-917. [PMID: 31526065 PMCID: PMC6758722 DOI: 10.1080/10717544.2019.1660435] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Although intraperitoneal chemotherapy (IPC) has been suggested as a promising method for the management of peritoneal dissemination (PD) of ovarian or colorectal cancers, the actual clinical use of this method has been restricted due to such problems as poor drug penetration into the tumor and high side effects. It is, therefore, necessary to develop new strategies to improve the efficacy of this approach. In the present work, a new strategy is proposed based on intraperitoneal (IP) injection of thermosensitive liposomal doxorubicin (TSL-Dox) with triggered release by mild hyperthermia induced by high intensity focused ultrasound (HIFU). A computational model is developed to evaluate the proposed drug delivery system. Results show an order of magnitude increase in drug penetration depth into the tumor compared to the conventional IP delivery. Furthermore, the effects of thermal conditions applied to the tumor, TSL size, tumor vessel permeability, and tumor size are investigated. Results indicate an improved efficiency of the drug delivery by expanding the heated region, yet, it increases the risk of unintentional TSL drug load release in the peritoneal cavity. Results also indicate that smaller TSLs have better treatment outcome. However, there is a significant reduction in treatment efficacy for TSLs with sizes smaller than the vessel wall pore size. Thus, tuning the size of TSL should be based on the tumor microvascular permeability. The simulation results suggest that the TSL-Dox delivery system in smaller tumors is far advantageous than larger ones. Results of our model can be used as guidelines for future preclinical studies.
Collapse
Affiliation(s)
- Mohsen Rezaeian
- Department of Mechanical Engineering, K. N. Toosi University of Technology , Tehran , Iran
| | - Amir Sedaghatkish
- Department of Mechanical Engineering, Isfahan University of Technology , Isfahan , Iran
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology , Tehran , Iran.,Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology , Tehran , Iran.,Department of Electrical and Computer Engineering, University of Waterloo , Waterloo , Canada.,Centre for Biotechnology and Bioengineering (CBB), University of Waterloo , Waterloo , Canada.,Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
40
|
Bok M, Zhao ZJ, Jeon S, Jeong JH, Lim E. Ultrasonically and Iontophoretically Enhanced Drug-Delivery System Based on Dissolving Microneedle Patches. Sci Rep 2020; 10:2027. [PMID: 32029808 PMCID: PMC7005184 DOI: 10.1038/s41598-020-58822-w] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 01/21/2020] [Indexed: 11/20/2022] Open
Abstract
A multifunctional system comprised of hyaluronic acid microneedles was developed as an effective transdermal delivery platform for rapid local delivery. The microneedles can regulate the filling amount on the tip, by controlling the concentration of hyaluronic acid solution. Ultrasonication induces dissolution of the HA microneedles via vibration of acoustic pressure, and AC iontophoresis improves the electrostatic force-driven diffusion of HA ions and rhodamine B. The effect of ultrasound on rhodamine release was analyzed in vitro using a gelatin hydrogel. The frequency and voltage dependence of the AC on the ion induction transfer was also evaluated experimentally. The results showed that the permeability of the material acts as a key material property. The delivery system based on ultrasonication and iontophoresis in microneedles increases permeation, thus resulting in shorter initial delivery time than that required by delivery systems based on passive or ultrasonication alone. This study highlights the significance of the combination between ultrasonic waves and iontophoresis for improving the efficiency of the microneedles, by shortening the reaction duration. We anticipate that this system can be extended to macromolecular and dependence delivery, based on drug response time.
Collapse
Affiliation(s)
- Moonjeong Bok
- Department of Science Education/Creative Convergent Manufacturing Engineering, Dankook University, Yongin, 16890, South Korea
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon, 34103, South Korea
| | - Zhi-Jun Zhao
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon, 34103, South Korea
| | - Sohee Jeon
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon, 34103, South Korea
| | - Jun-Ho Jeong
- Nano-Convergence Mechanical Systems Research Division, Korea Institute of Machinery and Materials, Daejeon, 34103, South Korea.
- Department of Nano Mechatronics, University of Science and Technology, Daejeon, 34103, South Korea.
| | - Eunju Lim
- Department of Science Education/Creative Convergent Manufacturing Engineering, Dankook University, Yongin, 16890, South Korea.
| |
Collapse
|
41
|
Abstract
Transport of drugs through the blood-brain barrier to the brain and the toxic effects of drugs on the healthy cells can limit the effectiveness of chemotherapeutic agents. In recent years, magnetic nanoparticles (MNPs) have received much attention as targeted therapeutic and diagnostic systems due to their simplicity, ease of preparation and ability to tailor their properties such as their composition, size, surface morphology, etc. for biomedical applications. MNPs are utilized in drug delivery, radio therapeutics, hyperthermia treatment, gene therapy, biotherapeutics and diagnostic imaging. The present review will address the challenges in brain tumor targeting and discuss the application and recent developments in brain tumor targeting using MNPs.
Collapse
|
42
|
Faridi P, Bossmann SH, Prakash P. Simulation-based design and characterization of a microwave applicator for MR-guided hyperthermia experimental studies in small animals. Biomed Phys Eng Express 2020; 6:015001. [PMID: 32999735 PMCID: PMC7521833 DOI: 10.1088/2057-1976/ab36dd] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Purpose The objective of this study was to design and characterize a 2.45 GHz microwave hyperthermia applicator for delivering hyperthermia in experimental small animals to 2 - 4 mm diameter targets located 1 - 3 mm from the skin surface, with minimal heating of the surrounding tissue, under 14.1 T MRI real-time monitoring and feedback control. Materials and methods An experimentally validated 3D computational model was employed to design and characterize a non-invasive directional water-cooled microwave hyperthermia applicator. We assessed the effects of: reflector geometry, monopole shape, cooling water temperature, and flow rate on spatial-temperature profiles. The system was integrated with real-time MR thermometry and feedback control to monitor and maintain temperature elevations in the range of 4 - 5 °C at 1 - 3 mm from the applicator surface. The quality of heating was quantified by determining the fraction of the target volume heated to the desired temperature, and the extent of heating in non-targeted regions. Results Model-predicted hyperthermic profiles were in good agreement with experimental measurements (Dice Similarity Coefficient of 0.95 - 0.99). Among the four considered criteria, a reflector aperture angle of 120 °, S-shaped monopole antenna with 0.6 mm displacement, and coolant flow rate of 150 ml/min were selected as the end result of the applicator design. The temperature of circulating water and input power were identified as free variables, allowing considerable flexibility in heating target sizes within varying distances from the applicator surface. 2 - 4 mm diameter targets positioned 1 - 3 mm from the applicator surface were heated to hyperthermic temperatures, with target coverage ratio ranging between 76 - 93 % and 11 - 26 % of non-targeted tissue heated. Conclusion We have designed an experimental platform for MR-guided hyperthermia, incorporating a microwave applicator integrated with temperature-based feedback control to heat deep-seated targets for experimental studies in small animals.
Collapse
Affiliation(s)
- Pegah Faridi
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Stefan H. Bossmann
- Department of Chemistry, Kansas State University, Manhattan, KS 66506, USA
| | - Punit Prakash
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
43
|
Thermosensitive Liposome-Mediated Drug Delivery in Chemotherapy: Mathematical Modelling for Spatio-temporal Drug Distribution and Model-Based Optimisation. Pharmaceutics 2019; 11:pharmaceutics11120637. [PMID: 31795486 PMCID: PMC6955700 DOI: 10.3390/pharmaceutics11120637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 11/17/2022] Open
Abstract
Thermosensitive liposome-mediated drug delivery has shown promising results in terms of improved therapeutic efficacy and reduced side effects compared to conventional chemotherapeutics. In order to facilitate our understanding of the transport mechanisms and their complex interplays in the drug delivery process, computational models have been developed to simulate the multiple steps involved in liposomal drug delivery to solid tumours. In this study we employ a multicompartmental model for drug-loaded thermosensitive liposomes, with an aim to identify the key transport parameters in determining therapeutic dosing and outcomes. The computational model allows us to not only examine the temporal and spatial variations of drug concentrations in the different compartments by utilising the tumour cord concept, but also assess the therapeutic efficacy and toxicity. In addition, the influences of key factors on systemic plasma concentration and intracellular concentration of the active drug are investigated; these include different chemotherapy drugs, release rate constants and heating duration. Our results show complex relationships between these factors and the predicted therapeutic outcome, making it difficult to identify the "best" parameter set. To overcome this challenge, a model-based optimisation method is proposed in an attempt to find a set of release rate constants and heating duration that can maximise intracellular drug concentration while minimising systemic drug concentration. Optimisation results reveal that under the operating conditions and ranges examined, the best outcome would be achieved with a low drug release rate at physiological temperature, combined with a moderate to high release rate at mild hyperthermia and 1 h heating after injection.
Collapse
|
44
|
Dehshahri A, Ashrafizadeh M, Ghasemipour Afshar E, Pardakhty A, Mandegary A, Mohammadinejad R, Sethi G. Topoisomerase inhibitors: Pharmacology and emerging nanoscale delivery systems. Pharmacol Res 2019; 151:104551. [PMID: 31743776 DOI: 10.1016/j.phrs.2019.104551] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/16/2019] [Indexed: 02/07/2023]
Abstract
Topoisomerase enzymes have shown unique roles in replication and transcription. These enzymes which were initially found in Escherichia coli have attracted considerable attention as target molecules for cancer therapy. Nowadays, there are several topoisomerase inhibitors in the market to treat or at least control the progression of cancer. However, significant toxicity, low solubility and poor pharmacokinetic properties have limited their wide application and these characteristics need to be improved. Nano-delivery systems have provided an opportunity to modify the intrinsic properties of molecules and also to transfer the toxic agent to the target tissues. These delivery systems leads to the re-introduction of existing molecules present in the market as novel therapeutic agents with different physicochemical and pharmacokinetic properties. This review focusses on a variety of nano-delivery vehicles used for the improvement of pharmacological properties of topoisomerase inhibitors and thus enabling their potential application as novel drugs in the market.
Collapse
Affiliation(s)
- Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Mandegary
- Physiology Research Center, Institute of Neuropharmacology, and Department of Toxicology & Pharmacology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| |
Collapse
|
45
|
Cheng CA, Chen W, Zhang L, Wu HH, Zink JI. A Responsive Mesoporous Silica Nanoparticle Platform for Magnetic Resonance Imaging-Guided High-Intensity Focused Ultrasound-Stimulated Cargo Delivery with Controllable Location, Time, and Dose. J Am Chem Soc 2019; 141:17670-17684. [PMID: 31604010 DOI: 10.1021/jacs.9b07591] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Magnetic resonance imaging (MRI) is an essential modality for clinical diagnosis, and MRI-guided high-intensity focused ultrasound (MRgHIFU) is a powerful technology for targeted therapy. Clinical applications of MRgHIFU primarily utilize hyperthermia and ablation to treat cancerous tissue, but for drug delivery applications thermal damage is undesirable. A biofriendly MRgHIFU-responsive mesoporous silica nanoparticle (MSN) platform that is stimulated within a physiological safe temperature range has been developed, reducing the possibility of thermal damage to the surrounding healthy tissues. Biocompatible polyethylene glycol (PEG) was employed to cap the pores of MSNs, and the release of cargo molecules by HIFU occurs without substantial temperature increase (∼4 °C). To visualize by MRI and measure the stimulated delivery in situ, a U.S. Food and Drug Administration (FDA)-approved gadolinium-based contrast agent, gadopentetate dimeglumine (Gd(DTPA)2-), was used as the imageable cargo. Taking advantage of the three-dimensional (3-D) imaging and targeting capabilities of MRgHIFU, the release of Gd(DTPA)2- stimulated by HIFU was pinpointed at the HIFU focal point in 3-D space in a tissue-mimicking gel phantom. The amount of Gd(DTPA)2- released was controlled by HIFU stimulation times and power levels. A positive correlation between the amount of Gd(DTPA)2- released and T1 was found. The MRgHIFU-stimulated cargo release was further imaged in a sample of ex vivo animal tissue. With this technology, the biodistribution of the nanocarriers can be tracked and the MRgHIFU-stimulated cargo release can be pinpointed, opening up an opportunity for future image-guided theranostic applications.
Collapse
Affiliation(s)
- Chi-An Cheng
- Department of Bioengineering , University of California Los Angeles , Los Angeles , California 90095 , United States.,California NanoSystems Institute , University of California Los Angeles , Los Angeles 90095 , California , United States
| | - Wei Chen
- Department of Chemistry & Biochemistry , University of California Los Angeles , Los Angeles , California 90095 , United States.,California NanoSystems Institute , University of California Los Angeles , Los Angeles 90095 , California , United States
| | - Le Zhang
- Department of Radiological Sciences, David Geffen School of Medicine , University of California Los Angeles , Los Angeles , California 90095 , United States
| | - Holden H Wu
- Department of Bioengineering , University of California Los Angeles , Los Angeles , California 90095 , United States.,Department of Radiological Sciences, David Geffen School of Medicine , University of California Los Angeles , Los Angeles , California 90095 , United States
| | - Jeffrey I Zink
- Department of Chemistry & Biochemistry , University of California Los Angeles , Los Angeles , California 90095 , United States.,California NanoSystems Institute , University of California Los Angeles , Los Angeles 90095 , California , United States
| |
Collapse
|
46
|
Prabhakar A, Banerjee R. Nanobubble Liposome Complexes for Diagnostic Imaging and Ultrasound-Triggered Drug Delivery in Cancers: A Theranostic Approach. ACS OMEGA 2019; 4:15567-15580. [PMID: 31572858 PMCID: PMC6761614 DOI: 10.1021/acsomega.9b01924] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/27/2019] [Indexed: 05/19/2023]
Abstract
The ability of ultrasound contrast agents to enhance the cell membrane permeability in response to an ultrasound pulse has unveiled avenues to facilitate the delivery of a higher intracellular payload at target sites. In light of the above, we report the development of submicron-sized (528.7 ± 31.7 nm) nanobubble-paclitaxel liposome (NB-PTXLp) complexes for ultrasound imaging and ultrasound responsive drug delivery in cancer cells. With a paclitaxel entrapment efficiency of 85.4 ± 4.39%, the 200 nm-sized liposomes tethered efficiently (conjugation efficiency ∼98.7 ± 0.14%) with the nanobubbles to form conjugates. Sonoporation of MiaPaCa-2 cells upon treatment with nanobubbles and ultrasound enhanced cellular permeability, resulting in 2.5-fold higher uptake of liposomes in comparison to only liposome treatment. This manifested into more than 300-fold higher anticancer activity of NB-PTXLps in the presence of ultrasound in MiaPaCa-2, Panc-1, MDA-MB-231, and AW-8507 cell lines, compared to commercial formulation ABRAXANE. Also, the NB-PTXLp conjugates were found to exhibit echogenicity comparable to the commercial ultrasound contrast agent SonoVue. In addition, the developed nanobubbles were found to exhibit more than 1 week echogenic stability as opposed to 6 h stability of the commercially available ultrasound contrast agent SonoVue. Hence, the NB-PTXLps developed herein could prove to be a promising and minimally invasive theranostic platform for cancer treatments in the future.
Collapse
|
47
|
Novoselova MV, Voronin DV, Abakumova TO, Demina PA, Petrov AV, Petrov VV, Zatsepin TS, Sukhorukov GB, Gorin DA. Focused ultrasound-mediated fluorescence of composite microcapsules loaded with magnetite nanoparticles: In vitro and in vivo study. Colloids Surf B Biointerfaces 2019; 181:680-687. [DOI: 10.1016/j.colsurfb.2019.06.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 01/02/2023]
|
48
|
Kang JH, Chon J, Kim YI, Lee HJ, Oh DW, Lee HG, Han CS, Kim DW, Park CW. Preparation and evaluation of tacrolimus-loaded thermosensitive solid lipid nanoparticles for improved dermal distribution. Int J Nanomedicine 2019; 14:5381-5396. [PMID: 31409994 PMCID: PMC6645695 DOI: 10.2147/ijn.s215153] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Tacrolimus (TCR), also known as FK-506, is a biopharmaceutics classification system (BCS) class II drug that is insoluble in water because of its high log P values. After dermal application, TCR remains in the stratum corneum and passes through the skin layers with difficulty. Purpose: The objectives of this study were to develop and evaluate solid lipid nanoparticles (SLNs) with thermosensitive properties to improve penetration and retention. Methods: We prepared TCR-loaded thermosensitive solid lipid nanoparticles (TCR-SLNs) with different types of surfactants on the shell of the particle, which conferred the advantages of enhancing skin permeation and distribution. We also characterized them from a physic point of view and performed in vitro and in vivo evaluations. Results: The TCR contained in the prepared TCR-SLN was in an amorphous state and entrapped in the particles with a high loading efficiency. The assessment of ex vivo skin penetration using excised rat dorsal skin showed that the TCR-SLNs penetrated to a deeper layer than the reference product (0.1% Protopic®). In addition, the in vivo skin penetration test demonstrated that TCR-SLNs delivered more drug into deeper skin layers than the reference product. FT-IR images also confirmed drug distribution of TCR-SLNs into deeper layers of the skin. Conclusion: These results revealed the potential application of thermosensitive SLNs for the delivery of difficult-to-permeate, poorly water-soluble drugs into deep skin layers.
Collapse
Affiliation(s)
- Ji-Hyun Kang
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Jinmann Chon
- School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Young-Il Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Hyo-Jung Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Dong-Won Oh
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Hong-Goo Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Chang-Soo Han
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | - Dong-Wook Kim
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju, Republic of Korea
| | - Chun-Woong Park
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
49
|
Bi H, Xue J, Jiang H, Gao S, Yang D, Fang Y, Shi K. Current developments in drug delivery with thermosensitive liposomes. Asian J Pharm Sci 2019; 14:365-379. [PMID: 32104466 PMCID: PMC7032122 DOI: 10.1016/j.ajps.2018.07.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 06/26/2018] [Accepted: 07/25/2018] [Indexed: 12/19/2022] Open
Abstract
Thermosensitive liposomes (TSLs) have been an important research area in the field of tumor targeted chemotherapy. Since the first TSLs appeared that using 1,2-dipalmitoyl-sn-glyce-ro-3-phosphocholine (DPPC) as the primary liposomal lipid, many studies have been done using this type of liposome from basic and practical aspects. While TSLs composed of DPPC enhance the cargo release near the phase transition temperature, it has been shown that many factors affect their temperature sensitivity. Thus numerous attempts have been undertaken to develop new TSLs for improving their thermal response performance. The main objective of this review is to introduce the development and recent update of innovative TSLs formulations, including combination of radiofrequency ablation (RFA), high-intensity focused ultrasound (HIFU), magnetic resonance imaging (MRI) and alternating magnetic field (AMF). In addition, various factors affecting the design of TSLs, such as lipid composition, surfactant, size and serum components are also discussed.
Collapse
Key Words
- (DPPC), 1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine
- (DPPGOG), 1,2-dipalmitoyl-sn-glycero-3-phosphoglyceroglycerol
- (DSPC), 1,2-distearoyl-sn-glycero-3-phosphocholine
- (DSPE-mPEG2000), 1,2-distearoyl-sn-glycero-3-phosphatiylethanol-amine-N-[methoxy(polyethyleneglycol)-2000]
- (LTSLs), lyso-lipid temperature sensitive liposomes
- (MPPC), 1-myristoyl-2-palmitoyl-sn-glycero-3-phosphatidylcholine
- (MSPC), 1-stearoyl-2-hydroxy-sn-glycero-3-phosphatidylcholine
- (P-lyso-PC), lysophosphatidylcholine
- (P188), 1-palmitoyl-2-stearoyl-sn-glycero-3-phosphatidylcholinex
- (P188), HO-(C2H4O)a-(C3H6O)b-(C2H4O)c-H, a=80, b=27, c=80
- Content release rate
- Drug delivery
- Hyperthermia
- Smart liposomes
- Thermosensitive liposomes
- Tumor chemotherapy
- fTSLs, fast release TSLs
- sTSLs, slow release TSLs
Collapse
Affiliation(s)
- Hongshu Bi
- Institute of New Drug Development, Liaoning Yaolian Pharmaceutical Co., Ltd., Benxi, Liaoning 117004, China
| | - Jianxiu Xue
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Hong Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Shan Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Dongjuan Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Yan Fang
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| | - Kai Shi
- School of Pharmacy, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, China
| |
Collapse
|
50
|
Reinišová L, Hermanová S, Pumera M. Micro/nanomachines: what is needed for them to become a real force in cancer therapy? NANOSCALE 2019; 11:6519-6532. [PMID: 30632584 DOI: 10.1039/c8nr08022d] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Conventional drug delivery systems face several issues in medical applications, such as cyto/genotoxicity and off-targeting. These issues are particularly significant for cancer therapeutics because many of the currently used systems are toxic in their free form. Self-propelled autonomous micro/nanomachines offer promising alternative drug delivery systems based on high cargo loading, fast autonomous movement, precise targeting and the on-demand release of therapeutics in vivo. With this unique set of properties, it is not surprising that they are receiving considerable research attention. However, much less is reported about the drawbacks that hinder their systemic in vivo application. In this review, a biomedical perspective is used to assess micro/nanomotor-based anticancer drug delivery systems reported to date. Advantages along with present issues are highlighted and recommendations which need to be considered to develop an effective biocompatible micro/nanomotor-based delivery system for cancer therapy are discussed.
Collapse
Affiliation(s)
- Lucie Reinišová
- Department of Polymers, Faculty of Chemical Technology, University of Chemistry and Technology Prague, Technická 5, 16628 Prague, Czech Republic
| | | | | |
Collapse
|