1
|
Zhao D, Zhang Y, Wang F, Kaewmanee R, Cui W, Wu T, Du Y. Drug-phospholipid conjugate nano-assembly for drug delivery. SMART MEDICINE 2024; 3:e20240053. [PMID: 39776594 PMCID: PMC11669785 DOI: 10.1002/smmd.20240053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025]
Abstract
Phospholipid-based liposomes are among the most successful nanodrug delivery systems in clinical use. However, these conventional liposomes present significant challenges including low drug-loading capacity and issues with drug leakage. Drug-phospholipid conjugates (DPCs) and their assemblies offer a promising strategy for addressing these limitations. In this review, we summarize recent advances in the design, synthesis, and application of DPCs for drug delivery. We begin by discussing the chemical backbone structures and various design strategies such as phosphate head embedding and mono-/bis-embedding in the sn-1/sn-2 positions. Furthermore, we highlight stimulus-responsive designs of DPCs and their applications in treating diseases such as cancer, inflammation, and malaria. Lastly, we explore future directions for DPCs development and their potential applications in drug delivery.
Collapse
Affiliation(s)
- Ding Zhao
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yixiang Zhang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fan Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Rames Kaewmanee
- Department of Materials ScienceFaculty of ScienceChulalongkorn UniversityBangkokThailand
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tianqi Wu
- Department of Radiation OncologyHuashan HospitalFudan UniversityShanghaiChina
| | - Yawei Du
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
2
|
Gao R, Lin P, Yang W, Fang Z, Gao C, Cheng B, Fang J, Yu W. Bio-Inspired Nanodelivery Platform: Platelet Membrane-Cloaked Genistein Nanosystem for Targeted Lung Cancer Therapy. Int J Nanomedicine 2024; 19:10455-10478. [PMID: 39430311 PMCID: PMC11491070 DOI: 10.2147/ijn.s479438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
Background Genistein (Gen), a natural polyphenolic compound, has emerged as a promising candidate for lung cancer treatment. However, the potential clinical application of Gen is limited due to its poor solubility, low bioavailability, and toxic side effects. To address these challenges, a biomimetic delivery platform with cell membranes derived from natural cells as carrier material was constructed. This innovative approach aims to facilitate targeted drug delivery and solve the problem of biocompatibility of synthetic materials. Methods First, the liposomes (LPs) loaded with Gen (LPs@Gen) was prepared using the ethanol injection method. Subsequently, PLTM-LPs@Gen was obtained through co-extrusion after mixing platelet membrane (PLTM) and LPs@Gen. Additionally, the biological and physicochemical properties of PLTM-LPs@Gen were investigated. Finally, the targeting ability, therapeutic efficacy, and safety of PLTM-LPs@Gen for lung cancer were evaluated using both a cell model and a tumor-bearing nude mouse model. Results The optimal preparation ratio for LPs@Gen was Gen: soybean lecithin: cholesterol: DSPE-PEG2000 (3:30:5:10, mass ratio), while the ideal fusion ratio of LPs@Gen and PLTM was 1:1. The particle size of PLTM-LPs@Gen was 108.33 ± 1.06 nm, and the encapsulation efficiency and drug loading were 94.29% and 3.09% respectively. Gen was released continuously and slowly from PLTM-LPs@Gen. Moreover, PLTM-LPs@Gen exhibited good stability within one week. The results of in vitro cellular uptake and in vivo distribution experiments indicated that the carrier material, PLTM-LPs, has the immune escape ability and tumor targeting ability. Consequently, it showed better therapeutic effects than free drugs and traditional LPs in vitro and in vivo tumor models. In addition, safety experiments demonstrated that PLTM-LPs@Gen possesses good biocompatibility. Conclusion Biomimetic nanomedicine provides a new strategy for the precision treatment of lung cancer in clinical practice.
Collapse
Affiliation(s)
- Rui Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, People’s Republic of China
| | - Peihong Lin
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, People’s Republic of China
| | - Wenjing Yang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, People’s Republic of China
| | - Zhengyu Fang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, People’s Republic of China
| | - Chunxiao Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, People’s Republic of China
| | - Bin Cheng
- Department of Traditional Chinese Medicine, Zhejiang Pharmaceutical University, Ningbo, 315500, People’s Republic of China
| | - Jie Fang
- Zhejiang Provincial Laboratory of Experimental Animal’s & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, 310013, People’s Republic of China
| | - Wenying Yu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, People’s Republic of China
| |
Collapse
|
3
|
Zou J. Site-specific delivery of cisplatin and paclitaxel mediated by liposomes: A promising approach in cancer chemotherapy. ENVIRONMENTAL RESEARCH 2023; 238:117111. [PMID: 37734579 DOI: 10.1016/j.envres.2023.117111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/25/2023] [Accepted: 09/09/2023] [Indexed: 09/23/2023]
Abstract
The site-specific delivery of drugs, especially anti-cancer drugs has been an interesting field for researchers and the reason is low accumulation of cytotoxic drugs in cancer cells. Although combination cancer therapy has been beneficial in providing cancer drug sensitivity, targeted delivery of drugs appears to be more efficient. One of the safe, biocompatible and efficient nano-scale delivery systems in anti-cancer drug delivery is liposomes. Their particle size is small and they have other properties such as adjustable physico-chemical properties, ease of functionalization and high entrapment efficiency. Cisplatin is a chemotherapy drug with clinical approval in patients, but its accumulation in cancer cells is low due to lack of targeted delivery and repeated administration results in resistance development. Gene and drug co-administration along with cisplatin/paclitaxel have resulted in increased sensitivity in tumor cells, but there is still space for more progress in cancer therapy. The delivery of cisplatin/paclitaxel by liposomes increases accumulation of drug in tumor cells and impairs activity of efflux pumps in promoting cytotoxicity. Moreover, phototherapy along with cisplatin/paclitaxel delivery can increase potential in tumor suppression. Smart nanoparticles including pH-sensitive nanoparticles provide site-specific delivery of cisplatin/paclitaxel. The functionalization of liposomes can be performed by ligands to increase targetability towards tumor cells in mediating site-specific delivery of cisplatin/paclitaxel. Finally, liposomes can mediate co-delivery of cisplatin/paclitaxel with drugs or genes in potentiating tumor suppression. Since drug resistance has caused therapy failure in cancer patients, and cisplatin/paclitaxel are among popular chemotherapy drugs, delivery of these drugs mediates targeted suppression of cancers and prevents development of drug resistance. Because of biocompatibility and safety of liposomes, they are currently used in clinical trials for treatment of cancer patients. In future, the optimal dose of using liposomes and optimal concentration of loading cisplatin/paclitaxel on liposomal nanocarriers in clinical trials should be determined.
Collapse
Affiliation(s)
- Jianyong Zou
- Department of Thoracic Surgery, The first Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, PR China.
| |
Collapse
|
4
|
Rama B, Ribeiro AJ. Role of nanotechnology in the prolonged release of drugs by the subcutaneous route. Expert Opin Drug Deliv 2023; 20:559-577. [PMID: 37305971 DOI: 10.1080/17425247.2023.2214362] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 05/11/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION Subcutaneous physiology is distinct from other parenteral routes that benefit the administration of prolonged-release formulations. A prolonged-release effect is particularly convenient for treating chronic diseases because it is associated with complex and often prolonged posologies. Therefore, drug-delivery systems focused on nanotechnology are proposed as alternatives that can overcome the limitations of current therapeutic regimens and improve therapeutic efficacy. AREAS COVERED This review presents an updated systematization of nanosystems, focusing on their applications in highly prevalent chronic diseases. Subcutaneous-delivered nanosystem-based therapies comprehensively summarize nanosystems, drugs, and diseases and their advantages, limitations, and strategies to increase their translation into clinical applications. An outline of the potential contribution of quality-by-design (QbD) and artificial intelligence (AI) to the pharmaceutical development of nanosystems is presented. EXPERT OPINION Although recent academic research and development (R&D) advances in the subcutaneous delivery of nanosystems have exhibited promising results, pharmaceutical industries and regulatory agencies need to catch up. The lack of standardized methodologies for analyzing in vitro data from nanosystems for subcutaneous administration and subsequent in vivo correlation limits their access to clinical trials. There is an urgent need for regulatory agencies to develop methods that faithfully mimic subcutaneous administration and specific guidelines for evaluating nanosystems.
Collapse
Affiliation(s)
- B Rama
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal
| | - A J Ribeiro
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal
- Genetics of Cognitive Disfunction, i3S, IBMC, Porto, Portugal
| |
Collapse
|
5
|
Paclitaxel-Loaded Lipid-Coated Magnetic Nanoparticles for Dual Chemo-Magnetic Hyperthermia Therapy of Melanoma. Pharmaceutics 2023; 15:pharmaceutics15030818. [PMID: 36986678 PMCID: PMC10055620 DOI: 10.3390/pharmaceutics15030818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/17/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Melanoma is the most aggressive and metastasis-prone form of skin cancer. Conventional therapies include chemotherapeutic agents, either as small molecules or carried by FDA-approved nanostructures. However, systemic toxicity and side effects still remain as major drawbacks. With the advancement of nanomedicine, new delivery strategies emerge at a regular pace, aiming to overcome these challenges. Stimulus-responsive drug delivery systems might considerably reduce systemic toxicity and side-effects by limiting drug release to the affected area. Herein, we report the development of paclitaxel-loaded lipid-coated manganese ferrite magnetic nanoparticles (PTX-LMNP) as magnetosomes synthetic analogs, envisaging the combined chemo-magnetic hyperthermia treatment of melanoma. PTX-LMNP physicochemical properties were verified, including their shape, size, crystallinity, FTIR spectrum, magnetization profile, and temperature profile under magnetic hyperthermia (MHT). Their diffusion in porcine ear skin (a model for human skin) was investigated after intradermal administration via fluorescence microscopy. Cumulative PTX release kinetics under different temperatures, either preceded or not by MHT, were assessed. Intrinsic cytotoxicity against B16F10 cells was determined via neutral red uptake assay after 48 h of incubation (long-term assay), as well as B16F10 cells viability after 1 h of incubation (short-term assay), followed by MHT. PTX-LMNP-mediated MHT triggers PTX release, allowing its thermal-modulated local delivery to diseased sites, within short timeframes. Moreover, half-maximal PTX inhibitory concentration (IC50) could be significantly reduced relatively to free PTX (142,500×) and Taxol® (340×). Therefore, the dual chemo-MHT therapy mediated by intratumorally injected PTX-LMNP stands out as a promising alternative to efficiently deliver PTX to melanoma cells, consequently reducing systemic side effects commonly associated with conventional chemotherapies.
Collapse
|
6
|
Nanoarchitectured assembly and surface of two-dimensional (2D) transition metal dichalcogenides (TMDCs) for cancer therapy. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
7
|
Rodrigues Arruda B, Mendes MGA, Freitas PGCD, Reis AVF, Lima T, Crisóstomo LCCF, Nogueira KAB, Pessoa C, Petrilli R, Eloy JO. Nanocarriers for delivery of taxanes: A review on physicochemical and biological aspects. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Updates on Responsive Drug Delivery Based on Liposome Vehicles for Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14102195. [PMID: 36297630 PMCID: PMC9608678 DOI: 10.3390/pharmaceutics14102195] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/20/2022] Open
Abstract
Liposomes are well-known nanoparticles with a non-toxic nature and the ability to incorporate both hydrophilic and hydrophobic drugs simultaneously. As modern drug delivery formulations are produced by emerging technologies, numerous advantages of liposomal drug delivery systems over conventional liposomes or free drug treatment of cancer have been reported. Recently, liposome nanocarriers have exhibited high drug loading capacity, drug protection, improved bioavailability, enhanced intercellular delivery, and better therapeutic effect because of resounding success in targeting delivery. The site targeting of smart responsive liposomes, achieved through changes in their physicochemical and morphological properties, allows for the controlled release of active compounds under certain endogenous or exogenous stimuli. In that way, the multifunctional and stimuli-responsive nanocarriers for the drug delivery of cancer therapeutics enhance the efficacy of treatment prevention and fighting over metastases, while limiting the systemic side effects on healthy tissues and organs. Since liposomes constitute promising nanocarriers for site-targeted and controlled anticancer drug release, this review focuses on the recent progress of smart liposome achievements for anticancer drug delivery applications.
Collapse
|
9
|
Abed HF, Abuwatfa WH, Husseini GA. Redox-Responsive Drug Delivery Systems: A Chemical Perspective. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3183. [PMID: 36144971 PMCID: PMC9503659 DOI: 10.3390/nano12183183] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 06/16/2023]
Abstract
With the widespread global impact of cancer on humans and the extensive side effects associated with current cancer treatments, a novel, effective, and safe treatment is needed. Redox-responsive drug delivery systems (DDSs) have emerged as a potential cancer treatment with minimal side effects and enhanced site-specific targeted delivery. This paper explores the physiological and biochemical nature of tumors that allow for redox-responsive drug delivery systems and reviews recent advances in the chemical composition and design of such systems. The five main redox-responsive chemical entities that are the focus of this paper are disulfide bonds, diselenide bonds, succinimide-thioether linkages, tetrasulfide bonds, and platin conjugates. Moreover, as disulfide bonds are the most commonly used entities, the review explored disulfide-containing liposomes, polymeric micelles, and nanogels. While various systems have been devised, further research is needed to advance redox-responsive drug delivery systems for cancer treatment clinical applications.
Collapse
Affiliation(s)
- Heba F. Abed
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad H. Abuwatfa
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
10
|
Sun L, Zhao P, Chen M, Leng J, Luan Y, Du B, Yang J, Yang Y, Rong R. Taxanes prodrug-based nanomedicines for cancer therapy. J Control Release 2022; 348:672-691. [PMID: 35691501 DOI: 10.1016/j.jconrel.2022.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/04/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022]
Abstract
Malignant tumor remains a huge threat to human health and chemotherapy still occupies an important place in clinical tumor treatment. As a kind of potent antimitotic agent, taxanes act as the first-line broad-spectrum cancer drug in clinical use. However, disadvantages such as prominent hydrophobicity, severe off-target toxicity or multidrug resistance lead to unsatisfactory therapeutic effects, which restricts its wider usage. The efficient delivery of taxanes is still quite a challenge despite the rapid developments in biomaterials and nanotechnology. Great progress has been made in prodrug-based nanomedicines (PNS) for cancer therapy due to their outstanding advantages such as high drug loading efficiency, low carrier induced immunogenicity, tumor stimuli-responsive drug release, combinational therapy and so on. Based on the numerous developments in this filed, this review summarized latest updates of taxanes prodrugs-based nanomedicines (TPNS), focusing on polymer-drug conjugate-based nanoformulations, small molecular prodrug-based self-assembled nanoparticles and prodrug-encapsulated nanosystems. In addition, the new trends of tumor stimuli-responsive TPNS were also discussed. Moreover, the future challenges of TPNS for clinical translation were highlighted. We here expect this review will inspire researchers to explore more practical taxanes prodrug-based nano-delivery systems for clinical use.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Menghan Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jiayi Leng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yixin Luan
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jia Yang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
11
|
Vilela JDMV, Moghassemi S, Dadashzadeh A, Dolmans MM, Azevedo RB, Amorim CA. Safety of Lavender Oil-Loaded Niosomes for In Vitro Culture and Biomedical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1999. [PMID: 35745338 PMCID: PMC9229298 DOI: 10.3390/nano12121999] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/30/2022] [Accepted: 06/09/2022] [Indexed: 12/22/2022]
Abstract
(1) Background: Essential oils have long been used as therapeutic agents. Lavender (Lavandula angustifolia) oil (LO) is an antispasmodic, anticonvulsant, relaxant, painkilling, and antimicrobial essential oil investigated as a natural substance for biomedical therapies. Nanoparticles have shown significant promise in improving drug delivery and efficacy. Considering these benefits, the aim of this study was to evaluate the toxicity of LO and lavender oil niosomes (LONs) in stem cells and myofibroblast models cultured in vitro. (2) Methods: Adipose tissue-derived stem cells and myometrial cells were cultured with LO or LONs at different concentrations (0, 0.016%, 0.031%, and 0.063%) and toxicity was evaluated with PrestoBlue™ and live/dead assay using calcein and ethidium homodimer. (3) Results: Cell viability was similar to controls in all groups, except in 0.063% LO for myometrial cells, which showed lower viability than the control medium. (4) Conclusion: These results suggest that both LO and LONs are safe for cell culture and may be used for pharmaceutical and biomedical therapies in future applications in regenerative medicine.
Collapse
Affiliation(s)
- Janice de M. V. Vilela
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (J.d.M.V.V.); (S.M.); (A.D.); (M.-M.D.)
| | - Saeid Moghassemi
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (J.d.M.V.V.); (S.M.); (A.D.); (M.-M.D.)
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (J.d.M.V.V.); (S.M.); (A.D.); (M.-M.D.)
| | - Marie-Madeleine Dolmans
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (J.d.M.V.V.); (S.M.); (A.D.); (M.-M.D.)
- Gynecology and Andrology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Ricardo B. Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil;
| | - Christiani A. Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium; (J.d.M.V.V.); (S.M.); (A.D.); (M.-M.D.)
| |
Collapse
|
12
|
Lou J, Schuster JA, Barrera FN, Best MD. ATP-Responsive Liposomes via Screening of Lipid Switches Designed to Undergo Conformational Changes upon Binding Phosphorylated Metabolites. J Am Chem Soc 2022; 144:3746-3756. [PMID: 35171601 DOI: 10.1021/jacs.2c00191] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Liposomal delivery vehicles can dramatically enhance drug transport. However, their clinical application requires enhanced control over content release at diseased sites. For this reason, triggered release strategies have been explored, although a limited toolbox of stimuli has thus far been developed. Here, we report a novel strategy for stimuli-responsive liposomes that release encapsulated contents in the presence of phosphorylated small molecules. Our formulation efforts culminated in selective cargo release driven by ATP, a universal energy source that is upregulated in diseases such as cancer. Specifically, we developed lipid switches 1a-b bearing two ZnDPA units designed to undergo substantial conformational changes upon ATP binding, thereby disrupting membrane packing and triggering the release of encapsulated contents. Dye leakage assays using the hydrophobic dye Nile red validated that ATP-driven release was selective over 11 similar phosphorylated metabolites, and release of the hydrophilic dye calcein was also achieved. Multiple alternative lipid switch structures were synthesized and studied (1c-d and 2), which provided insights into the structural features that render 1a-b selective toward ATP-driven release. Importantly, analysis of cellular delivery using fluorescence microscopy in conjunction with pharmacological ATP manipulation showed that liposome delivery was specific, as it increased upon intracellular ATP accumulation, and was inhibited by ATP downregulation. Our new approach shows strong prospects for enhancing the selectivity of release and payload delivery to diseased cells driven by metabolites such as ATP, providing an exciting new paradigm for controlled release.
Collapse
Affiliation(s)
- Jinchao Lou
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Jennifer A Schuster
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Francisco N Barrera
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Michael D Best
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| |
Collapse
|
13
|
Qualls ML, Sagar R, Lou J, Best MD. Demolish and Rebuild: Controlling Lipid Self-Assembly toward Triggered Release and Artificial Cells. J Phys Chem B 2021; 125:12918-12933. [PMID: 34792362 DOI: 10.1021/acs.jpcb.1c07406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The ability to modulate the structures of lipid membranes, predicated on our nuanced understanding of the properties that drive and alter lipid self-assembly, has opened up many exciting biological applications. In this Perspective, we focus on two endeavors in which the same principles are invoked to achieve completely opposite results. On one hand, controlled liposome decomposition enables triggered release of encapsulated cargo through the development of synthetic lipid switches that perturb lipid packing in the presence of disease-associated stimuli. In particular, recent approaches have utilized artificial lipid switches designed to undergo major conformational changes in response to a range of target conditions. On the other end of the spectrum, the ability to drive the in situ formation of lipid bilayer membranes from soluble precursors is an important component in the establishment of artificial cells. This work has culminated in chemoenzymatic strategies that enable lipid manufacturing from simple components. Herein, we describe recent advancements in these two unique undertakings that are linked by their reliance on common principles of lipid self-assembly.
Collapse
Affiliation(s)
- Megan L Qualls
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| | - Ruhani Sagar
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| | - Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, Tennessee 37996, United States
| |
Collapse
|
14
|
Stimuli-responsive nanoliposomes as prospective nanocarriers for targeted drug delivery. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102916] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
15
|
Hwang SR, Chakraborty K, An JM, Mondal J, Yoon HY, Lee YK. Pharmaceutical Aspects of Nanocarriers for Smart Anticancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13111875. [PMID: 34834290 PMCID: PMC8619450 DOI: 10.3390/pharmaceutics13111875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022] Open
Abstract
Drug delivery to tumor sites using nanotechnology has been demonstrated to overcome the drawbacks of conventional anticancer drugs. Altering the surface shape and geometry of nanocomposites alters their chemical properties, which can confer multiple attributes to nanocarriers for the treatment of cancer and their use as imaging agents for cancer diagnosis. However, heterogeneity and blood flow in human cancer limit the distribution of nanoparticles at the site of tumor tisues. For targeted delivery and controlled release of drug molecules in harsh tumor microenvironments, smart nanocarriers combined with various stimuli-responsive materials have been developed. In this review, we describe nanomaterials for smart anticancer therapy as well as their pharmaceutical aspects including pharmaceutical process, formulation, controlled drug release, drug targetability, and pharmacokinetic or pharmacodynamic profiles of smart nanocarriers. Inorganic or organic-inorganic hybrid nanoplatforms and the electrospinning process have also been briefly described here.
Collapse
Affiliation(s)
- Seung Rim Hwang
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea;
| | - Kushal Chakraborty
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju 27469, Korea;
| | - Jeong Man An
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea;
| | - Jagannath Mondal
- Department of Green Bio Engineering, Graduate School, Korea National University of Transportation, Chungju 27469, Korea;
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong 27909, Korea
| | - Hong Yeol Yoon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
| | - Yong-kyu Lee
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju 27469, Korea;
- Department of Green Bio Engineering, Graduate School, Korea National University of Transportation, Chungju 27469, Korea;
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea
- Correspondence: ; Tel.: +82-43-841-5224
| |
Collapse
|
16
|
Abd Elkodous M, Olojede SO, Morsi M, El-Sayyad GS. Nanomaterial-based drug delivery systems as promising carriers for patients with COVID-19. RSC Adv 2021; 11:26463-26480. [PMID: 35480012 PMCID: PMC9037715 DOI: 10.1039/d1ra04835j] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/27/2021] [Indexed: 01/09/2023] Open
Abstract
Once the World Health Organization (WHO) declared the COVID-19 outbreak to be pandemic, massive efforts have been launched by researchers around the globe to combat this emerging infectious disease. Here we review the most recent data on the novel SARS-CoV-2 pathogen. We analyzed its etiology, pathogenesis, diagnosis, prevention, and current medications. After that, we summarized the promising drug delivery application of nanomaterial-based systems. Their preparation routes, unique advantages over the traditional drug delivery routes and their toxicity though risk analysis were also covered. We also discussed in detail the mechanism of action for one example of drug-loaded nanomaterial drug delivery systems (Avigan-contained nano-emulsions). This review provides insights about employing nanomaterial-based drug delivery systems for the treatment of COVID-19 to increase the bioavailability of current drugs, reducing their toxicity, and to increase their efficiency.
Collapse
Affiliation(s)
- M Abd Elkodous
- Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology Toyohashi Aichi 441-8580 Japan
- Center for Nanotechnology (CNT), School of Engineering and Applied Sciences, Nile University Sheikh Zayed Giza 16453 Egypt
| | - S O Olojede
- Nanotechnology Platforms, Discipline of Clinical Anatomy, Nelson Mandela School of Medicine, University of KwaZulu-Natal Durban South Africa
| | - Mahmoud Morsi
- Faculty of Medicine, Menoufia University Menoufia Shebin El Kom Egypt
| | - Gharieb S El-Sayyad
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA) Cairo Egypt
- Chemical Engineering Department, Military Technical College (MTC) Egyptian Armed Forces Cairo Egypt
| |
Collapse
|
17
|
He W, Du Y, Wang T, Wang J, Cheng L, Li X. Redox responsive 7-ethyl-10-hydroxycamptothecin (SN38) lysophospholipid conjugate: synthesis, assembly and anticancer evaluation. Int J Pharm 2021; 606:120856. [PMID: 34229071 DOI: 10.1016/j.ijpharm.2021.120856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/01/2021] [Accepted: 07/01/2021] [Indexed: 12/25/2022]
Abstract
7-Ethyl-10-hydroxycamptothecin (SN38), a potent camptothecin derivative specifically targeting DNA topoisomerase I cleavage complexes, has shown great potential in the treatment of solid tumors. Because of its poor solubility and chemical and metabolic stability, the clinical application of SN38 is highly limited. To address these problems, a novel redox-responsive SN38 conjugate based liposomal formulation is developed in this report. First, SN38 was conjugated with lysophospholipid by using a cleavable disulfide bond linker. After that, the conjugate (SN38-SS-PC) was assembled into liposomes by thin film method. Dynamic lightscattering(DLS) characterization indicated that SN38-SS-PC liposomes possessed a narrow size distribution (172.8 ± 10.5 nm) and negative charged zeta potential (-8.9 ± 0.3 mV). The results of storage and physiological stabilities showed that SN38-SS-PC liposomes was stable under different conditions. More importantly, a reduction responsive release of parent drug SN38 was observed in the medium containing glutathione (GSH). In addition, SN38-SS-PC liposomes had a much more rapid cellular uptake behavior against cancer cells. The enhanced anti-cancer efficacy of SN38-SS-PC liposomes was further demonstrated by in vitro cytotoxicity assay against MCF-7 and A549 cells. Under in vivo evaluation in 4 T1 xenograft tumor model, SN38-SS-PC liposomes were observed to have lower systemic toxicity and higher tumor inhibition rate of 53.3% compared with the commercialized SN38 prodrug Irinotecan (Ir). In summary, SN38-SS-PC liposomes could be a promising redox responsive delivery system of SN38 for cancer therapy.
Collapse
Affiliation(s)
- Wei He
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yawei Du
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Tao Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ji Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Lei Cheng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
18
|
AlSawaftah N, Pitt WG, Husseini GA. Dual-Targeting and Stimuli-Triggered Liposomal Drug Delivery in Cancer Treatment. ACS Pharmacol Transl Sci 2021; 4:1028-1049. [PMID: 34151199 PMCID: PMC8205246 DOI: 10.1021/acsptsci.1c00066] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 12/31/2022]
Abstract
The delivery of chemotherapeutics to solid tumors using smart drug delivery systems (SDDSs) takes advantage of the unique physiology of tumors (i.e., disordered structure, leaky vasculature, abnormal extracellular matrix (ECM), and limited lymphatic drainage) to deliver anticancer drugs with reduced systemic side effects. Liposomes are the most promising of such SDDSs and have been well investigated for cancer therapy. To improve the specificity, bioavailability, and anticancer efficacy of liposomes at the diseased sites, other strategies such as targeting ligands and stimulus-sensitive liposomes have been developed. This review highlights relevant surface functionalization techniques and stimuli-mediated drug release for enhanced delivery of anticancer agents at tumor sites, with a special focus on dual functionalization and design of multistimuli responsive liposomes.
Collapse
Affiliation(s)
- Nour AlSawaftah
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, UAE
| | - William G. Pitt
- Chemical
Engineering Department, Brigham Young University, Provo, Utah 84602, United States
| | - Ghaleb A. Husseini
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, UAE
| |
Collapse
|
19
|
Stimulus-responsive liposomes for biomedical applications. Drug Discov Today 2021; 26:1794-1824. [PMID: 34058372 DOI: 10.1016/j.drudis.2021.05.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/01/2021] [Accepted: 05/17/2021] [Indexed: 02/08/2023]
Abstract
Liposomes are amphipathic lipidic supramolecular aggregates that are able to encapsulate and carry molecules of both hydrophilic and hydrophobic nature. They have been widely used as in vivo drug delivery systems for some time because they offer features such as synthetic flexibility, biodegradability, biocompatibility, low immunogenicity, and negligible toxicity. In recent years, the chemical modification of liposomes has paved the way to the development of smart liposome-based drug delivery systems, which are characterized by even more tunable and disease-directed features. In this review, we highlight the different types of chemical modification introduced to date, with a particular focus on internal stimuli-responsive liposomes and prodrug activation.
Collapse
|
20
|
Chen R, Wang Z, Wu S, Kuang X, Wang X, Yan G, Tang R. Chemosensitizing micelles self-assembled from amphiphilic TPGS-indomethacin twin drug for significantly synergetic multidrug resistance reversal. J Biomater Appl 2020; 35:994-1004. [PMID: 33283586 DOI: 10.1177/0885328220975177] [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: 11/17/2022]
Abstract
Vitamin E d-ɑ-tocopheryl poly(ethylene glycol) 1000 succinate (TPGS) and indomethacin (IDM) can reverse multidrug resistance (MDR) via inhibiting P-glycoprotein (P-gp) and multidrug resistance associated protein 1 (MRP1) respectively, but their drawbacks in physicochemical properties limit their clinical application. To overcome these defects and enhance MDR reversal, the amphiphilic TPGS-IDM twin drug was successfully synthesized via esterification, and could self-assemble into free and paclitaxel-loaded (PTX-loaded) micelles. The micelles exhibited lower CMC values (5.2 × 10-5 mg/mL), long-term stability in PBS (pH 7.4) for 7 days and SDS solution (5 mg/mL) for 3 days, and effective drug release at esterase/pH 5.0. Moreover, the micelles could down-regulate ATP levels and promote ROS production in MCF-7/ADR via the mitochondrial impairment, therefore achieving MDR reversal and cell apoptosis. Additionally, the PTX-loaded micelles could significantly inhibit the cell proliferation and promote apoptosis for MCF-7/ADR via the synergistic chemosensitizing effect of TPGS and IDM, and synergistic cytotoxic effect of TPGS and PTX. Thus, the chemosensitizing micelles self-assembled from amphiphilic TPGS-indomethacin twin drug have the great potentials for reversing MDR in clinical cancer therapy.
Collapse
Affiliation(s)
- Ran Chen
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, 12487Anhui University, Hefei, China *Equal contributors
| | - Zhexiang Wang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, 12487Anhui University, Hefei, China *Equal contributors
| | - Shuo Wu
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, 12487Anhui University, Hefei, China *Equal contributors
| | - Xingyu Kuang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, 12487Anhui University, Hefei, China *Equal contributors
| | - Xiu Wang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, 12487Anhui University, Hefei, China *Equal contributors
| | - Guoqing Yan
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, 12487Anhui University, Hefei, China *Equal contributors
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, 12487Anhui University, Hefei, China *Equal contributors
| |
Collapse
|
21
|
Almeida B, Nag OK, Rogers KE, Delehanty JB. Recent Progress in Bioconjugation Strategies for Liposome-Mediated Drug Delivery. Molecules 2020; 25:E5672. [PMID: 33271886 PMCID: PMC7730700 DOI: 10.3390/molecules25235672] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
In nanoparticle (NP)-mediated drug delivery, liposomes are the most widely used drug carrier, and the only NP system currently approved by the FDA for clinical use, owing to their advantageous physicochemical properties and excellent biocompatibility. Recent advances in liposome technology have been focused on bioconjugation strategies to improve drug loading, targeting, and overall efficacy. In this review, we highlight recent literature reports (covering the last five years) focused on bioconjugation strategies for the enhancement of liposome-mediated drug delivery. These advances encompass the improvement of drug loading/incorporation and the specific targeting of liposomes to the site of interest/drug action. We conclude with a section highlighting the role of bioconjugation strategies in liposome systems currently being evaluated for clinical use and a forward-looking discussion of the field of liposomal drug delivery.
Collapse
Affiliation(s)
- Bethany Almeida
- American Society for Engineering Education, Washington, DC 20036, USA;
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
| | - Okhil K. Nag
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
| | - Katherine E. Rogers
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
- Fischell Department of Bioengineering, 2330 Kim Engineering Building, University of Maryland, College Park, MD 20742, USA
| | - James B. Delehanty
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
| |
Collapse
|
22
|
Yu J, Liu Y, Zhou S, Wang Y, Wang Y. Stimuli-responsive phospholipid-drug conjugates (PDCs)-based nanovesicles for drug delivery and theranostics. Int J Pharm 2020; 590:119920. [PMID: 33002539 DOI: 10.1016/j.ijpharm.2020.119920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 01/07/2023]
Abstract
Liposomes represent one of the most successful nano-drug delivery systems among enormous nano-carriers. Although great progress has been made in conventional liposomes, the emerging shortcomings still impair the therapeutic index. The proposal of stimuli-responsive phospholipid-drug conjugates (PDCs)-based nanovesicles solves the challenges that conventional liposomes are faced with, showing great potential for cancer diagnosis and therapy. Herein, we intend to overview the current progress and unique advantages of stimuli-responsive PDCs-based nanovesicles. First, the challenges of conventional liposomes and the development of PDCs-based nanovesicles are summarized. Next, the stimuli-responsive elements used in current stimuli-responsive PDCs-based nanovesicles are outlined. Then, the unique superiorities of stimuli-responsive PDCs-based nanovesicles for drug delivery and theranostics are highlighted in detail. Finally, the future opportunities and challenges of stimuli-responsive PDCs-based nanovesicles for clinical translation are put forward.
Collapse
Affiliation(s)
- Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Ying Liu
- National Institute for Food and Drug Control, Beijing 102629, China
| | - Shuang Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yingli Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
23
|
Chen S, Song Z, Feng R. Recent Development of Copolymeric Nano-Drug Delivery System for Paclitaxel. Anticancer Agents Med Chem 2020; 20:2169-2189. [PMID: 32682385 DOI: 10.2174/1871520620666200719001038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/25/2020] [Accepted: 06/25/2020] [Indexed: 11/22/2022]
Abstract
Background:
Paclitaxel (PTX) has been clinically used for several years due to its good therapeutic
effect against cancers. Its poor water-solubility, non-selectivity, high cytotoxicity to normal tissue and worse
pharmacokinetic property limit its clinical application.
Objective:
To review the recent progress on the PTX delivery systems.
Methods:
In recent years, the copolymeric nano-drug delivery systems for PTX are broadly studied. It mainly
includes micelles, nanoparticles, liposomes, complexes, prodrugs and hydrogels, etc. They were developed or
further modified with target molecules to investigate the release behavior, targeting to tissues, pharmacokinetic
property, anticancer activities and bio-safety of PTX. In the review, we will describe and discuss the recent
progress on the nano-drug delivery system for PTX since 2011.
Results:
The water-solubility, selective delivery to cancers, tissue toxicity, controlled release and pharmacokinetic
property of PTX are improved by its encapsulation into the nano-drug delivery systems. In addition, its
activities against cancer are also comparable or high when compared with the commercial formulation.
Conclusion:
Encapsulating PTX into nano-drug carriers should be helpful to reduce its toxicity to human, keeping
or enhancing its activity and improving its pharmacokinetic property.
Collapse
Affiliation(s)
- Shiyu Chen
- School of Biological Science and Technology, University of Jinan, No. 336 West Road of Nanxinzhuang, Jinan 250022, Shandong Province, China
| | - Zhimei Song
- School of Biological Science and Technology, University of Jinan, No. 336 West Road of Nanxinzhuang, Jinan 250022, Shandong Province, China
| | - Runliang Feng
- School of Biological Science and Technology, University of Jinan, No. 336 West Road of Nanxinzhuang, Jinan 250022, Shandong Province, China
| |
Collapse
|
24
|
Lou J, Best MD. Strategies for altering lipid self-assembly to trigger liposome cargo release. Chem Phys Lipids 2020; 232:104966. [PMID: 32888913 DOI: 10.1016/j.chemphyslip.2020.104966] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 01/21/2023]
Abstract
While liposomes have proven to be effective drug delivery nanocarriers, their therapeutic attributes could be improved through the development of clinically viable triggered release strategies in which encapsulated drug contents could be selectively released at the sites of diseased cells. As such, a significant amount of research has been reported involving the development of stimuli-responsive liposomes and a broad range of strategies have been explored for driving content release. These have included the introduction of trigger groups at either the lipid headgroup or within the acyl chains that alter lipid self-assembly properties of known lipids as well as the rational design of lipid analogs programed to undergo conformational changes induced by events such as binding interactions. This review article describes advances in the design of stimuli-responsive liposome strategies with an eye towards emerging trends in the field.
Collapse
Affiliation(s)
- Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Dr, Knoxville, TN, 37996, USA
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Dr, Knoxville, TN, 37996, USA.
| |
Collapse
|
25
|
Ma Y, Mou Q, Yan D, Zhu X. Engineering small molecule nanodrugs to overcome barriers for cancer therapy. VIEW 2020. [DOI: 10.1002/viw.20200062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Yuan Ma
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| | - Quanbing Mou
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
26
|
Kumar K, Kumar Shyamlal BR, Verma R, Kondaiah P, Chaudhary S. Reduction-Triggered Doxorubicin Delivery by Self-Assembled Nanospheres of Lipoylated Caffeine. ChemMedChem 2020; 15:733-737. [PMID: 32162419 DOI: 10.1002/cmdc.202000070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/06/2020] [Indexed: 01/09/2023]
Abstract
This study reports a new amphiphilic bioconjugate (CAFF-LA) derived from the lipoylation of a hydroxyethyl derivative of caffeine. In water, CAFF-LA self-assembles into nanospheres with an average size of 155 nm, as evidenced from dynamic light scattering and electron microscopy studies. The nanospheres are stable in serum and could be disintegrated upon exposure to the reducing environment of dithiothreitol (DTT; 10 mM) and glutathione (GSH; 10 mM). These nanospheres easily encapsulate the chemotherapy medication, doxorubicin (DOX), and demonstrate an efficacious transport into doxorubicin-resistant cervical cancer (HeLa) cells, wherein a marked induction in apoptosis and significantly lower IC50 have been observed when compared to that of free drug. The in vitro assessment of cell viability and hemocompatibility present these nanospheres as potentially safe and efficient intracellular reduction stimulus-responsive drug-delivery vehicles.
Collapse
Affiliation(s)
- Krishan Kumar
- Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur, 302017, India
| | - Bharti Rajesh Kumar Shyamlal
- Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur, 302017, India
| | - Rajbala Verma
- Department of Zoology, University of Rajasthan, Jaipur, 302004, India
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Sandeep Chaudhary
- Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur, 302017, India
| |
Collapse
|
27
|
Li N, Guo W, Li Y, Zuo H, Zhang H, Wang Z, Zhao Y, Yang F, Ren G, Zhang S. Construction and anti-tumor activities of disulfide-linked docetaxel-dihydroartemisinin nanoconjugates. Colloids Surf B Biointerfaces 2020; 191:111018. [PMID: 32304917 DOI: 10.1016/j.colsurfb.2020.111018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 12/19/2022]
Abstract
Co-delivery of anti-tumor agents with outstanding stimulus-triggered drug release in tumor cells, especially with the aid of nanotechnology, provided the possibility to enhance delivery efficiency for targeting tumor cells and antitumor efficacy. In this paper, docetaxel-dihydroartemisinin nanoconjugates linked by disulfide bond were designed to increase co-delivery and anti-tumor efficacy. Docetaxel and dihydroartemisinin were synthesized using two-step reaction and furtherly assembled to nanoconjugates. Nanoprescription was optimized to evaluate its physicochemical properties. In vitro anti-tumor activities of nanoformulation were assessed by MTT. The flow cytometry was adopted to analyze cell apoptosis and cell cycle arrest. The wound healing assay was used to evaluate antimigratory-property. In vivo pharmacokinetic and pharmacodynamic studies were investigated in rats and 4T1 bearing Balb/c mice model after intravenous injection, respectively. The chemical structure of conjugate was confirmed. The prepared nanoparticles possessed uniform size distribution (172.10 ± 1.70 nm, PDI 0.05 ± 0.01), was stable during storage period, sustained release profiles and sensitive reduction responsiveness. MTT assay indicated that the toxicity of nanoconjugates was slightly weak. Flow cytometry studies showed that nanoconjugates could promote early apoptosis significantly and mainly arose from G0/G1 phase. The wound healing assay provided an obvious antimetastatic potential of nanoparticles in 4T1 cells. The result of pharmacokinetic study suggested that nanoconjugates exhibited higher exposure levels. In vivo pharmacodynamic research showed that mice treated with docetaxel-dihydroartemisinin nanoconjugates had lower systemic toxicity and higher survival ratio than those of control groups. This potential of nanoconjugates was developed as a novel nanoplateform to treat tumor.
Collapse
Affiliation(s)
- Ning Li
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenju Guo
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yujie Li
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hengtong Zuo
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huihui Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhaoyun Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongdan Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fan Yang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guolian Ren
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Shuqiu Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
28
|
Martin-Serrano Á, Gómez R, Ortega P, de la Mata FJ. Nanosystems as Vehicles for the Delivery of Antimicrobial Peptides (AMPs). Pharmaceutics 2019; 11:E448. [PMID: 31480680 PMCID: PMC6781550 DOI: 10.3390/pharmaceutics11090448] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/24/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022] Open
Abstract
Recently, antimicrobial peptides (AMPs), also called host defence peptides (HDPs), are attracting great interest, as they are a highly viable alternative in the search of new approaches to the resistance presented by bacteria against antibiotics in infectious diseases. However, due to their nature, they present a series of disadvantages such as low bioavailability, easy degradability by proteases, or low solubility, among others, which limits their use as antimicrobial agents. For all these reasons, the use of vehicles for the delivery of AMPs, such as polymers, nanoparticles, micelles, carbon nanotubes, dendrimers, and other types of systems, allows the use of AMPs as a real alternative to treatment with antibiotics.
Collapse
Affiliation(s)
- Ángela Martin-Serrano
- Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), University of Alcalá, 28805 Madrid, Spain
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
| | - Rafael Gómez
- Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), University of Alcalá, 28805 Madrid, Spain
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Paula Ortega
- Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), University of Alcalá, 28805 Madrid, Spain.
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.
| | - F Javier de la Mata
- Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), University of Alcalá, 28805 Madrid, Spain.
- Institute Ramón y Cajal for Health Research (IRYCIS), 28034 Madrid, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.
| |
Collapse
|
29
|
Snell A, Neupane KR, McCorkle JR, Fu X, Moonschi FH, Caudill EB, Kolesar J, Richards CI. Cell-Derived Vesicles for in Vitro and in Vivo Targeted Therapeutic Delivery. ACS OMEGA 2019; 4:12657-12664. [PMID: 31460386 PMCID: PMC6681979 DOI: 10.1021/acsomega.9b01353] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/11/2019] [Indexed: 06/01/2023]
Abstract
Efficient delivery of therapeutics across the cell membrane to the interior of the cell remains a challenge both in vitro and in vivo. Here, we demonstrate that vesicles derived from cellular membranes can be efficiently loaded with cargo that can then be delivered to the interior of the cell. These vesicles demonstrated cell-targeting specificity as well as the ability to deliver a wide range of different cargos. We utilized this approach to deliver both lipophilic and hydrophilic cargos including therapeutics and DNA in vitro. We further demonstrated in vivo targeting and delivery using fluorescently labeled vesicles to target tumor xenografts in an animal. Cell-derived vesicles can be generated in high yields and are easily loaded with a variety of cargos. The ability of these vesicles to specifically target the same cell type from which they originated provides an efficient means of delivering cargo, such as therapeutics, both in vitro and in vivo.
Collapse
Affiliation(s)
- Aaron
A. Snell
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Khaga R. Neupane
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - J. Robert McCorkle
- Markey Cancer Center and Department of Pharmacy Practice &
Science, College
of Pharmacy, University of Kentucky, Lexington, Kentucky 40508, United States
| | - Xu Fu
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Faruk H. Moonschi
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Elizabeth B. Caudill
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Jill Kolesar
- Markey Cancer Center and Department of Pharmacy Practice &
Science, College
of Pharmacy, University of Kentucky, Lexington, Kentucky 40508, United States
| | | |
Collapse
|
30
|
Du Y, He W, Zhou W, Li X. Disulfide phosphatidylcholines: alternative phospholipids for the preparation of functional liposomes. Chem Commun (Camb) 2019; 55:8434-8437. [DOI: 10.1039/c9cc03571k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Disulfide phosphatidylcholine-based liposomes can be triggered and completely destroyed in a reductive tumor microenvironment resulting in a fast release of encapsulated drugs.
Collapse
Affiliation(s)
- Yawei Du
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing
- China
| | - Wei He
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing
- China
| | - Wenya Zhou
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing
- China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing
- China
| |
Collapse
|