1
|
Abawi A, Trunfio-Sfarghiu AM, Thomann C, Petiot E, Lollo G, Granjon T, Girard-Egrot A, Maniti O. Tailor-made vincristine-liposomes for tumor targeting. Biochimie 2024; 227:35-46. [PMID: 39094823 DOI: 10.1016/j.biochi.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
To ensure selective targeting based on membrane fluidity and physico-chemical compatibility between the biological membrane of the target cell and the lipid membrane of the liposomes carriers. Lipid-based carriers as liposomes with varying membrane fluidities were designed for delivering vincristine, an anti-tumor compound derived from Madagascar's periwinkle. Liposomes, loaded with vincristine, were tested on prostate, colon, and breast cancer cell lines alongside non-tumor controls. Results showed that vincristine-loaded liposomes with fluid membranes significantly decreased the viability of cancer cell lines compared to controls. Confocal microscopy revealed the intracellular release of vincristine, evidenced by disrupted mitosis-specific labeling of actin filaments in metastatic prostate cell lines. This highlights the crucial role of membrane fluidity in the development of lipid-based drug carriers, offering a promising and cost-effective option for targeting cancer cells as an alternative to conventional strategies.
Collapse
Affiliation(s)
- Ariana Abawi
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | | | - Céline Thomann
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Emma Petiot
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Giovanna Lollo
- Laboratoire D'Automatique, de Génie des Procédés et de Génie Pharmaceutique, LAGEPP UMR 5007, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Thierry Granjon
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Agnès Girard-Egrot
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| | - Ofelia Maniti
- Institute of Molecular and Supramolecular Chemistry and Biochemistry, ICBMS UMR 5246, Univ. Lyon, University Lyon 1, CNRS, 69622, Lyon, France.
| |
Collapse
|
2
|
Pande S. Factors affecting response variables with emphasis on drug release and loading for optimization of liposomes. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:334-344. [PMID: 38833335 DOI: 10.1080/21691401.2024.2360634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Drug delivery through Liposomes has shown tremendous potential in terms of the therapeutic application of nanoparticles. There are several drug-loaded liposomal formulations approved for clinical use that help mitigate harmful effects of life-threatening diseases. Developments in the field of liposomal formulations and drug delivery have made it possible for clinicians and researchers to find therapeutic solutions for complicated medical conditions. A key aspect in the development of drug-loaded liposomes is a careful review of optimization techniques to improve the overall formulation stability and efficacy. Optimization studies help in improving/modulating the various properties of drug-loaded liposomes and are vital for the development of this class of delivery systems. A comprehensive overview of the various process variables and factors involved in the optimization of drug-loaded liposomes is presented in this review. The influence of different independent variables on drug release and loading properties with the application of a statistical experimental design is also explained in this article.
Collapse
Affiliation(s)
- Shantanu Pande
- Drug Product Technical Services, Wave Life Sciences, MA, USA
| |
Collapse
|
3
|
Dejeu IL, Vicaș LG, Marian E, Ganea M, Frenț OD, Maghiar PB, Bodea FI, Dejeu GE. Innovative Approaches to Enhancing the Biomedical Properties of Liposomes. Pharmaceutics 2024; 16:1525. [PMID: 39771504 PMCID: PMC11728823 DOI: 10.3390/pharmaceutics16121525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 10/31/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Liposomes represent a promising class of drug delivery systems that enhance the therapeutic efficacy and safety of various pharmaceutical agents. Also, they offer numerous advantages compared to traditional drug delivery methods, including targeted delivery to specific sites, controlled release, and fewer side effects. This review meticulously examines the methodologies employed in the preparation and characterization of liposomal formulations. With the rising incidence of adverse drug reactions, there is a pressing need for innovative delivery strategies that prioritize selectivity, specificity, and safety. Nanomedicine promises to revolutionize diagnostics and treatments, addressing current limitations and improving disease management, including cancer, which remains a major global health challenge. This paper aims to conduct a comprehensive study on the interest of biomedical research regarding nanotechnology and its implications for further applications.
Collapse
Affiliation(s)
- Ioana Lavinia Dejeu
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Laura Grațiela Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Mariana Ganea
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Olimpia Daniela Frenț
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania; (I.L.D.); (E.M.); (M.G.); (O.D.F.)
| | - Paula Bianca Maghiar
- Doctoral School of Biomedical Science, University of Oradea, 1 University Street, 410087 Oradea, Romania; (P.B.M.); (F.I.B.)
| | - Flaviu Ionut Bodea
- Doctoral School of Biomedical Science, University of Oradea, 1 University Street, 410087 Oradea, Romania; (P.B.M.); (F.I.B.)
| | - George Emanuiel Dejeu
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 Piata 1 Decembrie Street, 410073 Oradea, Romania;
| |
Collapse
|
4
|
Zhao Y, Wang ZM, Song D, Chen M, Xu Q. Rational design of lipid nanoparticles: overcoming physiological barriers for selective intracellular mRNA delivery. Curr Opin Chem Biol 2024; 81:102499. [PMID: 38996568 PMCID: PMC11323194 DOI: 10.1016/j.cbpa.2024.102499] [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: 03/13/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 07/14/2024]
Abstract
This review introduces the typical delivery process of messenger RNA (mRNA) nanomedicines and concludes that the delivery involves a at least four-step SCER cascade and that high efficiency at every step is critical to guarantee high overall therapeutic outcomes. This SCER cascade process includes selective organ-targeting delivery, cellular uptake, endosomal escape, and cytosolic mRNA release. Lipid nanoparticles (LNPs) have emerged as a state-of-the-art vehicle for in vivo mRNA delivery. The review emphasizes the importance of LNPs in achieving selective, efficient, and safe mRNA delivery. The discussion then extends to the technical and clinical considerations of LNPs, detailing the roles of individual components in the SCER cascade process, especially ionizable lipids and helper phospholipids. The review aims to provide an updated overview of LNP-based mRNA delivery, outlining recent innovations and addressing challenges while exploring future developments for clinical translation over the next decade.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Zeyu Morgan Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Donghui Song
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Mengting Chen
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
5
|
Zeng M, Wu B, Wei W, Jiang Z, Li P, Quan Y, Hu X. Disulfiram: A novel repurposed drug for cancer therapy. Chin Med J (Engl) 2024; 137:1389-1398. [PMID: 38275022 PMCID: PMC11188872 DOI: 10.1097/cm9.0000000000002909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Indexed: 01/27/2024] Open
Abstract
ABSTRACT Cancer is a major global health issue. Effective therapeutic strategies can prolong patients' survival and reduce the costs of treatment. Drug repurposing, which identifies new therapeutic uses for approved drugs, is a promising approach with the advantages of reducing research costs, shortening development time, and increasing efficiency and safety. Disulfiram (DSF), a Food and Drug Administration (FDA)-approved drug used to treat chronic alcoholism, has a great potential as an anticancer drug by targeting diverse human malignancies. Several studies show the antitumor effects of DSF, particularly the combination of DSF and copper (DSF/Cu), on a wide range of cancers such as glioblastoma (GBM), breast cancer, liver cancer, pancreatic cancer, and melanoma. In this review, we summarize the antitumor mechanisms of DSF/Cu, including induction of intracellular reactive oxygen species (ROS) and various cell death signaling pathways, and inhibition of proteasome activity, as well as inhibition of nuclear factor-kappa B (NF-κB) signaling. Furthermore, we highlight the ability of DSF/Cu to target cancer stem cells (CSCs), which provides a new approach to prevent tumor recurrence and metastasis. Strikingly, DSF/Cu inhibits several molecular targets associated with drug resistance, and therefore it is becoming a novel option to increase the sensitivity of chemo-resistant and radio-resistant patients. Studies of DSF/Cu may shed light on its improved application to clinical tumor treatment.
Collapse
Affiliation(s)
- Min Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Baibei Wu
- The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Wenjie Wei
- Institute of Biochemistry of Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zihan Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Peiqiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yuanting Quan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaobo Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
6
|
Shariat Razavi F, Kouchak M, Sistani Karampour N, Mahdavinia M, Nazari Khorasgani Z, Rezaie A, Rahbar N. AS1411aptamer conjugated liposomes for targeted delivery of arsenic trioxide in mouse xenograft model of melanoma cancer. J Liposome Res 2024; 34:288-302. [PMID: 37843918 DOI: 10.1080/08982104.2023.2271046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Development of AS1411aptamer-conjugated liposomes for targeted delivery of arsenic trioxide is the primary goal of this study. AS1411aptamer was used as ligand to target nucleolin, which is highly expressed on the surface of melanoma cancer cells. The targeted liposomes were constructed by the thin film method, and arsenic trioxide was loaded as cobalt (II) hydrogen arsenite (CHA) to increase the loading efficiency and stability of the liposomes. The liposomal structure was characterized by Fourier Transform Infrared Spectroscopy (FT-IR) and field emission scanning electron microscopy (FESEM). In addition, particle sizes and zeta potential of the CHA-loaded liposomes (CHAL) and aptamer-functionalized CHA-loaded liposomes (AP-CHAL) were determined. In vitro cytotoxicity of CHAL and AP-CHAL were evaluated using MTT assay in murine melanoma (B16) and mouse embryonic fibroblast (MEF) cell lines. The encapsulation efficiency of CHAL and AP-CHAL was reported as 60.2 ± 6.5% and 58.7 ± 4.2%, respectively. In vivo antitumor activity of CHAL and AP-CHAL in the B16 tumor-xenograft mouse model was dramatically observed. All mice of both groups survived until the end of treatment and showed body weight gain. The tumor protrusion completely disappeared in 50% of the mice in these groups. Furthermore, histopathology studies demonstrated that CHAL and AP-CHAL did not induce significant toxicity in healthy mice tissues. However, unlike the CHAL group, which showed an initial increase in tumor volume, a specific antitumor effect was observed in the AP-CHAL group from the beginning of treatment. The results showed that AP-CHAL can be used as an effective drug delivery system with high potential in the treatment of solid tumors.
Collapse
Affiliation(s)
- Fatemeh Shariat Razavi
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Neda Sistani Karampour
- Marine Pharmaceutical Science Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Nazari Khorasgani
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Annahita Rezaie
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Nadereh Rahbar
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
7
|
Moghimipour E, Handali S. Functionalized liposomes as a potential drug delivery systems for colon cancer treatment: A systematic review. Int J Biol Macromol 2024; 269:132023. [PMID: 38697444 DOI: 10.1016/j.ijbiomac.2024.132023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Colon cancer is one of the lethal diseases in the world with approximately 700,000 fatalities annually. Nowadays, due to the side effects of existing methods in the treatment of colon cancer such as radiotherapy and chemotherapy, the use of targeted nanocarriers in cancer treatment has received wide attention, and among them, especially liposomes have been studied a lot. Based on this, anti-tumor drugs hidden in targeted active liposomes can selectively act on cancer cells. In this systematic review, the use of various ligands such as folic acid, transferrin, aptamer, hyaluronic acid and cRGD for active targeting of liposomes to achieve improved drug delivery to colon cancer cells has been reviewed. The original articles published in English in the databases of Science Direct, PubMed and Google scholar from 2012 to 2022 were reviewed. From the total of 26,256 published articles, 19 studies met the inclusion criteria. The results of in vitro and in vivo studies have revealed that targeted liposomes lead to increasing the efficacy of anti-cancer agents on colon cancer cells with reducing side effects compared to free drugs and non-targeted liposomes. To the best of our knowledge, this is the first systematic review showing promising results for improvement treatment of colon cancer using targeted liposomes.
Collapse
Affiliation(s)
- Eskandar Moghimipour
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Handali
- Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Hu J, Pang J, Chen L, Li Y, Gan N, Pan Q, Wu D. Photoresponsive Azobenzene Nanocluster-Modified Liposomes: Mechanism Analysis Combining Experiments and Simulations. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:9761-9774. [PMID: 38663878 DOI: 10.1021/acs.langmuir.4c00787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Stimuli-responsive behaviors and controlled release in liposomes are pivotal in nanomedicine. To this end, we present an approach using a photoresponsive azobenzene nanocluster (AzDmpNC), prepared from azobenzene compounds through melting and aggregation. When integrated with liposomes, they form photoresponsive vesicles. The morphology and association with liposomes were investigated by using transmission electron microscopy. Liposomes loaded with calcein exhibited a 9.58% increased release after UV exposure. To gain insights into the underlying processes and elucidate the mechanisms involved. The molecular dynamic simulations based on the reactive force field and all-atom force field were employed to analyze the aggregation of isomers into nanoclusters and their impacts on phospholipid membranes, respectively. The results indicate that the nanoclusters primarily aggregate through π-π and T-stacking forces. The force density inside the cis-isomer of AzDmpNC formed after photoisomerization is lower, leading to its easier dispersion, rapid diffusion, and penetration into the membrane, disrupting the densification.
Collapse
Affiliation(s)
- Jie Hu
- School of Mechanical Engineering, Chengdu University, Chengdu 610106, China
| | - Jingtao Pang
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Lijuan Chen
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yilin Li
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Na Gan
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | | |
Collapse
|
9
|
Duché G, Sanderson JM. The Chemical Reactivity of Membrane Lipids. Chem Rev 2024; 124:3284-3330. [PMID: 38498932 PMCID: PMC10979411 DOI: 10.1021/acs.chemrev.3c00608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
It is well-known that aqueous dispersions of phospholipids spontaneously assemble into bilayer structures. These structures have numerous applications across chemistry and materials science and form the fundamental structural unit of the biological membrane. The particular environment of the lipid bilayer, with a water-poor low dielectric core surrounded by a more polar and better hydrated interfacial region, gives the membrane particular biophysical and physicochemical properties and presents a unique environment for chemical reactions to occur. Many different types of molecule spanning a range of sizes, from dissolved gases through small organics to proteins, are able to interact with membranes and promote chemical changes to lipids that subsequently affect the physicochemical properties of the bilayer. This Review describes the chemical reactivity exhibited by lipids in their membrane form, with an emphasis on conditions where the lipids are well hydrated in the form of bilayers. Key topics include the following: lytic reactions of glyceryl esters, including hydrolysis, aminolysis, and transesterification; oxidation reactions of alkenes in unsaturated fatty acids and sterols, including autoxidation and oxidation by singlet oxygen; reactivity of headgroups, particularly with reactive carbonyl species; and E/Z isomerization of alkenes. The consequences of reactivity for biological activity and biophysical properties are also discussed.
Collapse
Affiliation(s)
- Genevieve Duché
- Génie
Enzimatique et Cellulaire, Université
Technologique de Compiègne, Compiègne 60200, France
| | - John M Sanderson
- Chemistry
Department, Durham University, Durham DH1 3LE, United Kingdom
| |
Collapse
|
10
|
Xia Y, Cao K, Jia R, Chen X, Wu Y, Wang Y, Cheng Z, Xia H, Xu Y, Xie Z. Tetramethylpyrazine-loaded liposomes surrounded by hydrogel based on sodium alginate and chitosan as a multifunctional drug delivery System for treatment of atopic dermatitis. Eur J Pharm Sci 2024; 193:106680. [PMID: 38128842 DOI: 10.1016/j.ejps.2023.106680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/24/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Tetramethylpyrazine (TMP) has low bioavailability due to its fast metabolism and short half-life, which is not conducive to transdermal treatment of atopic dermatitis (AD). Therefore, in this study, TMP was encapsulated into liposomes (Lip) by film dispersion method, and then the surface of Lip was modified by sodium alginate (ALG) and chitosan (CS). The tetramethylpyrazine-loaded liposomes in sodium alginate chitosan hydrogel called T-Lip-AC hydrogel. In vitro experiments, we found that T-Lip-AC hydrogel not only had the antibacterial effect of CS, but also enhanced the anti-inflammatory and antioxidant effects of TMP. In addition, T-Lip-AC hydrogel could also provide a moist healing environment for AD dry skin and produce better skin permeability, and can also achieve sustained drug release, which is conducive to the treatment of AD. The lesions induced by 1-chloro-2,4-dinitrobenzene were used as the AD lesions model to test the therapeutic effect of the T-Lip-AC hydrogel on AD in vivo. The studies have showed that T-Lip-AC hydrogel could effectively promote wound healing. Therefore, we have developed a T-Lip-AC hydrogel as multifunctional hydrogel drug delivery system, which could become an effective, safe and novel alternative treatment method for treating AD.
Collapse
Affiliation(s)
- Ying Xia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Keang Cao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ruoyang Jia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Xue Chen
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yang Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yu Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Zhiqing Cheng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Hongmei Xia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Yinxiang Xu
- Zhaoke (Hefei) Pharmaceutical Co., Ltd., Hefei, 230088, China
| | - Zili Xie
- Anhui Institute for Food and Drug Control, Hefei, 230051, China
| |
Collapse
|
11
|
Qi QR, Tian H, Yue BS, Zhai BT, Zhao F. Research Progress of SN38 Drug Delivery System in Cancer Treatment. Int J Nanomedicine 2024; 19:945-964. [PMID: 38293612 PMCID: PMC10826519 DOI: 10.2147/ijn.s435407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/22/2023] [Indexed: 02/01/2024] Open
Abstract
The active metabolite of irinotecan (CPT-11), 7-ethyl-10-hydroxycamptothecin (SN38), is 100-1000 times more active than CPT-11 and has shown inhibitory effects on a range of cancer cells, including those from the rectal, small cell lung, breast, esophageal, uterine, and ovarian malignancies. Despite SN38's potent anticancer properties, its hydrophobicity and pH instability have caused substantial side effects and anticancer activity loss, which make it difficult to use in clinical settings. To solve the above problems, the construction of SN38-based drug delivery systems is one of the most feasible methods to improve drug solubility, enhance drug stability, increase drug targeting ability, improve drug bioavailability, enhance therapeutic efficacy and reduce adverse drug reactions. Therefore, based on the targeting mechanism of drug delivery systems, this paper reviews SN38 drug delivery systems, including polymeric micelles, liposomal nanoparticles, polymeric nanoparticles, protein nanoparticles, conjugated drug delivery systems targeted by aptamers and ligands, antibody-drug couplings, magnetic targeting, photosensitive targeting, redox-sensitive and multi-stimulus-responsive drug delivery systems, and co-loaded drug delivery systems. The focus of this review is on nanocarrier-based SN38 drug delivery systems. We hope to provide a reference for the clinical translation and application of novel SN38 medications.
Collapse
Affiliation(s)
- Qing-rui Qi
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Huan Tian
- Xi’an Hospital of Traditional Chinese Medicine, Xi’an, 710021, People’s Republic of China
| | - Bao-sen Yue
- Xi’an Hospital of Traditional Chinese Medicine, Xi’an, 710021, People’s Republic of China
| | - Bing-tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Feng Zhao
- Xi’an Hospital of Traditional Chinese Medicine, Xi’an, 710021, People’s Republic of China
| |
Collapse
|
12
|
Pande S. Liposomes for drug delivery: review of vesicular composition, factors affecting drug release and drug loading in liposomes. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:428-440. [PMID: 37594208 DOI: 10.1080/21691401.2023.2247036] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
Liposomes are considered among the most versatile and advanced nanoparticle delivery systems used to target drugs to specific cells and tissues. Structurally, liposomes are sphere-like vesicles of phospholipid molecules that are surrounded by equal number of aqueous compartments. The spherical shell encapsulates an aqueous interior which contains substances such as peptides and proteins, hormones, enzymes, antibiotics, antifungal and anticancer agents. This structural property of liposomes makes it an important nano-carrier for drug delivery. Extrusion is one of the most frequently used technique for preparing monodisperse uni-lamellar liposomes as the technique is used to control vesicle size. The process involves passage of lipid suspension through polycarbonate membrane with a fixed pore size to produce vesicles with a diameter near the pore size of the membrane used in preparing them. An advantage of this technique is that there is no need to remove the organic solvent or detergent from the final preparation. This review focuses on composition of liposome formulation with special emphasis on factors affecting drug release and drug-loading.
Collapse
Affiliation(s)
- Shantanu Pande
- Drug Product Technical Services, Wave Life Sciences, Lexington, MA, USA
| |
Collapse
|
13
|
Giordani S, Marassi V, Zattoni A, Roda B, Reschiglian P. Liposomes characterization for market approval as pharmaceutical products: Analytical methods, guidelines and standardized protocols. J Pharm Biomed Anal 2023; 236:115751. [PMID: 37778202 DOI: 10.1016/j.jpba.2023.115751] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/13/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Liposomes are nano-sized lipid-based vesicles widely studied for their drug delivery capabilities. Compared to standard carries they exhibit better properties such as improved site-targeting and drug release, protection of drugs from degradation and clearance, and lower toxic side effects. At present, scientific literature is rich of studies regarding liposomes-based systems, while 14 types of liposomal products have been authorized to the market by EMA and FDA and many others have been approved by national agencies. Although the interest in nanodevices and nanomedicine has steadily increased in the last two decades the development of documentation regulating and standardizing all the phases of their development and quality control still suffers from major inadequacy due to the intrinsic complexity of nano-systems characterization. Many generic documents (Type 1) discussing guidelines for the study of nano-systems (lipidic and not) have been proposed while there is a lack of robust and standardized methods (Type 2 documents). As a result, a widespread of different techniques, approaches and methodologies are being used, generating results of variable quality and hard to compare with each other. Additionally, such documents are often subject to updates and rewriting further complicating the topic. Within this context the aim of this work is focused on bridging the gap in liposome characterization: the most recent standardized methodologies suitable for liposomes characterization are here reported (with the corresponding Type 2 documents) and revised in a short and pragmatical way focused on providing the reader with a practical background of the state of the art. In particular, this paper will put the accent on the methodologies developed to evaluate the main critical quality attributes (CQAs) necessary for liposomes market approval.
Collapse
Affiliation(s)
- Stefano Giordani
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy
| | - Valentina Marassi
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy.
| | - Andrea Zattoni
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy
| | - Barbara Roda
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy.
| | - Pierluigi Reschiglian
- Department of Chemistry "Giacomo Ciamician", University of Bologna, 40126 Bologna, Italy; byFlow srl, 40129 Bologna, Italy
| |
Collapse
|
14
|
Bonechi C, Mahdizadeh FF, Talarico L, Pepi S, Tamasi G, Leone G, Consumi M, Donati A, Magnani A. Liposomal Encapsulation of Citicoline for Ocular Drug Delivery. Int J Mol Sci 2023; 24:16864. [PMID: 38069187 PMCID: PMC10706088 DOI: 10.3390/ijms242316864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Glaucoma represents a group of neurodegenerative diseases characterized by optic nerve damage and the slowly progressive death of retinal ganglion cells. Glaucoma is considered the second leading cause of irreversible blindness worldwide. Pharmaceutical treatment of glaucoma is critical because of the properties of the ocular barrier that limit the penetration of drugs, resulting in lower systemic bioavailability. This behavior causes the need of frequent drug administration, which leads to deposition of concentrated solutions on the eye, causing toxic effects and cellular damage to the eye. To overcome these drawbacks, novel drug-delivery systems, such as liposomes, can play an important role in improving the therapeutic efficacy of antiglaucomatous drugs. In this work, liposomes were synthesized to improve various aspects, such as ocular barrier penetration, bioavailability, sustained release of the drug, targeting of the tissue, and reduction in intraocular pressure. Citicoline (CDP-choline; cytidine 5'-diphosphocholine) is an important intermediate in the biosynthesis of cell membrane phospholipids, with neuroprotective and neuroenhancement properties, and it was used in the treatment on retinal function and neural conduction in the visual pathways of glaucoma patients. In this study, citicoline was loaded into the 1,2-dioleoyl-sn-glycerol-3-phosphocholine and cholesterol liposomal carrier to enhance its therapeutic effect. The citicoline encapsulation efficiency, drug release, and size analysis of the different liposome systems were investigated using dynamic light scattering, nuclear magnetic resonance, infrared spectroscopy, and ToF-SIMS experiments.
Collapse
Affiliation(s)
- Claudia Bonechi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (F.F.M.); (L.T.); (S.P.); (G.T.); (G.L.); (M.C.)
- Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Fariba Fahmideh Mahdizadeh
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (F.F.M.); (L.T.); (S.P.); (G.T.); (G.L.); (M.C.)
| | - Luigi Talarico
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (F.F.M.); (L.T.); (S.P.); (G.T.); (G.L.); (M.C.)
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti 9, 50121 Firenze, Italy
| | - Simone Pepi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (F.F.M.); (L.T.); (S.P.); (G.T.); (G.L.); (M.C.)
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti 9, 50121 Firenze, Italy
| | - Gabriella Tamasi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (F.F.M.); (L.T.); (S.P.); (G.T.); (G.L.); (M.C.)
- Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Gemma Leone
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (F.F.M.); (L.T.); (S.P.); (G.T.); (G.L.); (M.C.)
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti 9, 50121 Firenze, Italy
| | - Marco Consumi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (F.F.M.); (L.T.); (S.P.); (G.T.); (G.L.); (M.C.)
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti 9, 50121 Firenze, Italy
| | - Alessandro Donati
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (F.F.M.); (L.T.); (S.P.); (G.T.); (G.L.); (M.C.)
- Centre for Colloid and Surface Science (CSGI), University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Agnese Magnani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (C.B.); (F.F.M.); (L.T.); (S.P.); (G.T.); (G.L.); (M.C.)
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Via G. Giusti 9, 50121 Firenze, Italy
| |
Collapse
|
15
|
Lu X, Cheng H, Xu Q, Tan X. Encapsulation of STING Agonist cGAMP with Folic Acid-Conjugated Liposomes Significantly Enhances Antitumor Pharmacodynamic Effect. Cancer Biother Radiopharm 2023; 38:543-557. [PMID: 33719535 DOI: 10.1089/cbr.2020.4085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background: 2',3'-cGAMP (2',3'-cyclic AMP-GMP) has been reported as an agonist of the STING (stimulator of interferon genes) signaling pathway. However, cGAMP has poor membrane permeability and can be hydrolyzed by ectonucleotide pyrophosphatase/phosphodiesterase (ENPP1), limiting its ability to activate the STING-IRF3 pathway. This study aimed to investigate that the folate-targeted liposomal cGAMP could overcome the defects of free cGAMP to enhance the antitumor effect. Materials and Methods: cGAMP was encapsulated in PEGylated folic acid-targeted liposomes to construct a carrier-delivered formulation. The particle size and morphology were detected by dynamic light scattering and transmission electron microscopy. The sustained-release ability was measured by drug release and pharmacokinetics. Animal models were applied to evaluate the tumor inhibition efficiency in vivo. Flow cytometry, enzyme-linked immunosorbent assay, and real-time polymerase chain reaction were used to detect the expression of immune cells, secreted cytokines, and target genes. The activation of the STING-IRF3 pathway was evaluated by immunofluorescence. Results: Physical characters of liposomes revealed that the prepared liposomes were stable in neutral humoral environments and released more internal drugs in acidic tumor tissues. Systemic therapy with liposomes on Colorectal 26 tumor-bearing mice in vivo effectively inhibited tumor growth via stimulating the expression of CD8+ T cells and reversed the immunosuppressed tumor microenvironment (TME). Conclusions: The study suggests that the folic acid-targeted cGAMP-loaded liposomes deliver drugs to the TME to enhance the STING agonist activity, improving the efficiency of tumor therapy via the cGAMP-STING-IRF3 pathway.
Collapse
Affiliation(s)
- Xing Lu
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hao Cheng
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qiming Xu
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiangshi Tan
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Xu H, Wu L, Xue Y, Yang T, Xiong T, Wang C, He S, Sun H, Cao Z, Liu J, Wang S, Li Z, Naeem A, Yin X, Zhang J. Advances in Structure Pharmaceutics from Discovery to Evaluation and Design. Mol Pharm 2023; 20:4404-4429. [PMID: 37552597 DOI: 10.1021/acs.molpharmaceut.3c00514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Drug delivery systems (DDSs) play an important role in delivering active pharmaceutical ingredients (APIs) to targeted sites with a predesigned release pattern. The chemical and biological properties of APIs and excipients have been extensively studied for their contribution to DDS quality and effectiveness; however, the structural characteristics of DDSs have not been adequately explored. Structure pharmaceutics involves the study of the structure of DDSs, especially the three-dimensional (3D) structures, and its interaction with the physiological and pathological structure of organisms, possibly influencing their release kinetics and targeting abilities. A systematic overview of the structures of a variety of dosage forms, such as tablets, granules, pellets, microspheres, powders, and nanoparticles, is presented. Moreover, the influence of structures on the release and targeting capability of DDSs has also been discussed, especially the in vitro and in vivo release correlation and the structure-based organ- and tumor-targeting capabilities of particles with different structures. Additionally, an in-depth discussion is provided regarding the application of structural strategies in the DDSs design and evaluation. Furthermore, some of the most frequently used characterization techniques in structure pharmaceutics are briefly described along with their potential future applications.
Collapse
Affiliation(s)
- Huipeng Xu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Wu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Ministry of Education, Yantai University, Yantai 264005, China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yanling Xue
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Ting Yang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ting Xiong
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Caifen Wang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siyu He
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongyu Sun
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zeying Cao
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Liu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siwen Wang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhe Li
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Abid Naeem
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xianzhen Yin
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 201602, China
| | - Jiwen Zhang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, No.2 Tiantan Xili, Beijing 100050, China
| |
Collapse
|
17
|
Koroleva M. Multicompartment colloid systems with lipid and polymer membranes for biomedical applications. Phys Chem Chem Phys 2023; 25:21836-21859. [PMID: 37565484 DOI: 10.1039/d3cp01984e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Multicompartment structures have the potential for biomedical applications because they can act as multifunctional systems and provide simultaneous delivery of drugs and diagnostics agents of different types. Moreover, some of them mimic biological cells to some extent with organelles as separate sub-compartments. This article analyses multicompartment colloidal structures with smaller sub-units covered with lipid or polymer membranes that provide additional protection for the encapsulated substances. Vesosomes with small vesicles encapsulated in the inner pools of larger liposomes are the most studied systems to date. Dendrimer molecules are enclosed by a lipid bilayer shell in dendrosomes. Capsosomes, polymersomes-in-polymer capsules, and cubosomes-in-polymer capsules are composed of sub-compartments encapsulated within closed multilayer polymer membranes. Janus or Cerberus emulsions contain droplets composed of two or three phases: immiscible oils in O/W emulsions and aqueous polymer or salt solutions that are separated into two or three phases and form connected droplets in W/O emulsions. In more cases, the external surface of engulfed droplets in Janus or Cerberus emulsions is covered with a lipid or polymer monolayer. eLiposomes with emulsion droplets encapsulated into a bilayer shell have been given little attention so far, but they have very great prospects. In addition to nanoemulsion droplets, solid lipid nanoparticles, nanostructured lipid carriers and inorganic nanoparticles can be loaded into eLiposomes. Molecular engineering of the external membrane allows the creation of ligand-targeted and stimuli-responsive multifunctional systems. As a result, the efficacy of drug delivery can be significantly enhanced.
Collapse
Affiliation(s)
- Marina Koroleva
- Mendeleev University of Chemical Technology, Miusskaya sq. 9, Moscow 125047.
| |
Collapse
|
18
|
Emerging potential of 5-Fluorouracil-loaded chitosan nanoparticles in cancer therapy. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
19
|
Lim C, Shin Y, Kang K, Husni P, Lee D, Lee S, Choi HG, Lee ES, Youn YS, Oh KT. Effects of PEG-Linker Chain Length of Folate-Linked Liposomal Formulations on Targeting Ability and Antitumor Activity of Encapsulated Drug. Int J Nanomedicine 2023; 18:1615-1630. [PMID: 37020691 PMCID: PMC10069508 DOI: 10.2147/ijn.s402418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Introduction Ligand-conjugated liposomes are promising for the treatment of specific receptor-overexpressing cancers. However, previous studies have shown inconsistent results because of the varying properties of the ligand, presence of a polyethylene glycol (PEG) coating on the liposome, length of the linker, and density of the ligand. Methods Here, we prepared PEGylated liposomes using PEG-linkers of various lengths conjugated with folate and evaluated the effect of the PEG-linker length on the nanoparticle distribution and pharmacological efficacy of the encapsulated drug both in vitro and in vivo. Results When folate was conjugated to the liposome surface, the cellular uptake efficiency in folate receptor overexpressed KB cells dramatically increased compared to that of the normal liposome. However, when comparing the effect of the PEG-linker length in vitro, no significant difference between the formulations was observed. In contrast, the level of tumor accumulation of particles in vivo significantly increased when the length of the PEG-linker was increased. The tumor size was reduced by >40% in the Dox/FL-10K-treated group compared to that in the Dox/FL-2K- or 5K-treated groups. Discussion Our study suggests that as the length of PEG-linker increases, the tumor-targeting ability can be enhanced under in vivo conditions, which can lead to an increase in the antitumor activity of the encapsulated drug.
Collapse
Affiliation(s)
- Chaemin Lim
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yuseon Shin
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Kioh Kang
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Patihul Husni
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dayoon Lee
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sehwa Lee
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Hanyang University, Ansan, 15588, South Korea
| | - Eun Seong Lee
- Department of Biotechnology, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, 06974, Republic of Korea
- Correspondence: Kyung Taek Oh, College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul, 06974, Republic of Korea, Tel +82-2-824-5617, Email
| |
Collapse
|
20
|
Draguet F, Bouland C, Dubois N, Bron D, Meuleman N, Stamatopoulos B, Lagneaux L. Potential of Mesenchymal Stromal Cell-Derived Extracellular Vesicles as Natural Nanocarriers: Concise Review. Pharmaceutics 2023; 15:pharmaceutics15020558. [PMID: 36839879 PMCID: PMC9964668 DOI: 10.3390/pharmaceutics15020558] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/29/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Intercellular communication, through direct and indirect cell contact, is mandatory in multicellular organisms. These last years, the microenvironment, and in particular, transfer by extracellular vesicles (EVs), has emerged as a new communication mechanism. Different biological fluids and cell types are common sources of EVs. EVs play different roles, acting as signalosomes, biomarkers, and therapeutic agents. As therapeutic agents, MSC-derived EVs display numerous advantages: they are biocompatible, non-immunogenic, and stable in circulation, and they are able to cross biological barriers. Furthermore, EVs have a great potential for drug delivery. Different EV isolation protocols and loading methods have been tested and compared. Published and ongoing clinical trials, and numerous preclinical studies indicate that EVs are safe and well tolerated. Moreover, the latest studies suggest their applications as nanocarriers. The current review will describe the potential for MSC-derived EVs as drug delivery systems (DDS) in disease treatment, and their advantages. Thereafter, we will outline the different EV isolation methods and loading techniques, and analyze relevant preclinical studies. Finally, we will describe ongoing and published clinical studies. These elements will outline the benefits of MSC-derived EV DDS over several aspects.
Collapse
Affiliation(s)
- Florian Draguet
- Laboratory of Clinical Cell Therapy (LCCT), Jules Bordet Institute, Université Libre de Bruxelles (ULB), 90 Rue Meylemeersch, 1070 Brussels, Belgium
- Correspondence:
| | - Cyril Bouland
- Laboratory of Clinical Cell Therapy (LCCT), Jules Bordet Institute, Université Libre de Bruxelles (ULB), 90 Rue Meylemeersch, 1070 Brussels, Belgium
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, 322 Rue Haute, 1000 Brussels, Belgium
- Department of Maxillofacial and Reconstructive Surgery, Grand Hôpital de Charleroi, 3 Grand’Rue, 6000 Charleroi, Belgium
| | - Nathan Dubois
- Laboratory of Clinical Cell Therapy (LCCT), Jules Bordet Institute, Université Libre de Bruxelles (ULB), 90 Rue Meylemeersch, 1070 Brussels, Belgium
| | - Dominique Bron
- Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles (ULB), 90 Rue Meylemeersch, 1070 Brussels, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy (LCCT), Jules Bordet Institute, Université Libre de Bruxelles (ULB), 90 Rue Meylemeersch, 1070 Brussels, Belgium
- Department of Haematology, Jules Bordet Institute, Université Libre de Bruxelles (ULB), 90 Rue Meylemeersch, 1070 Brussels, Belgium
- Medicine Faculty, Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| | - Basile Stamatopoulos
- Laboratory of Clinical Cell Therapy (LCCT), Jules Bordet Institute, Université Libre de Bruxelles (ULB), 90 Rue Meylemeersch, 1070 Brussels, Belgium
- Medicine Faculty, Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy (LCCT), Jules Bordet Institute, Université Libre de Bruxelles (ULB), 90 Rue Meylemeersch, 1070 Brussels, Belgium
| |
Collapse
|
21
|
Zafar MN, Abuwatfa WH, Husseini GA. Acoustically-Activated Liposomal Nanocarriers to Mitigate the Side Effects of Conventional Chemotherapy with a Focus on Emulsion-Liposomes. Pharmaceutics 2023; 15:421. [PMID: 36839744 PMCID: PMC9963571 DOI: 10.3390/pharmaceutics15020421] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
To improve currently available cancer treatments, nanomaterials are employed as smart drug delivery vehicles that can be engineered to locally target cancer cells and respond to stimuli. Nanocarriers can entrap chemotherapeutic drugs and deliver them to the diseased site, reducing the side effects associated with the systemic administration of conventional anticancer drugs. Upon accumulation in the tumor cells, the nanocarriers need to be potentiated to release their therapeutic cargo. Stimulation can be through endogenous or exogenous modalities, such as temperature, electromagnetic irradiation, ultrasound (US), pH, or enzymes. This review discusses the acoustic stimulation of different sonosensitive liposomal formulations. Emulsion liposomes, or eLiposomes, are liposomes encapsulating phase-changing nanoemulsion droplets, which promote acoustic droplet vaporization (ADV) upon sonication. This gives eLiposomes the advantage of delivering the encapsulated drug at low intensities and short exposure times relative to liposomes. Other formulations integrating microbubbles and nanobubbles are also discussed.
Collapse
Affiliation(s)
- Mah Noor Zafar
- Biomedical Engineering Program, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad H. Abuwatfa
- Department of Chemical and Biological 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 and Biological 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
|
22
|
Gharoonpour A, Simiyari D, Yousefzadeh A, Badragheh F, Rahmati M. Autophagy modulation in breast cancer utilizing nanomaterials and nanoparticles. Front Oncol 2023; 13:1150492. [PMID: 37213283 PMCID: PMC10196239 DOI: 10.3389/fonc.2023.1150492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/19/2023] [Indexed: 05/23/2023] Open
Abstract
Autophagy regenerates cellular nutrients, recycles metabolites, and maintains hemostasis through multistep signaling pathways, in conjunction with lysosomal degradation mechanisms. In tumor cells, autophagy has been shown to play a dual role as both tumor suppressor and tumor promoter, leading to the discovery of new therapeutic strategies for cancer. Therefore, regulation of autophagy is essential during cancer progression. In this regard, the use of nanoparticles (NPs) is a promising technique in the clinic to modulate autophagy pathways. Here, we summarized the importance of breast cancer worldwide, and we discussed its classification, current treatment strategies, and the strengths and weaknesses of available treatments. We have also described the application of NPs and nanocarriers (NCs) in breast cancer treatment and their capability to modulate autophagy. Then the advantages and disadvantaged of NPs in cancer therapy along with future applications will be disscussed. The purpose of this review is to provide up-to-date information on NPs used in breast cancer treatment and their impacts on autophagy pathways for researchers.
Collapse
|
23
|
Upadhyay P, Agarwal S, Upadhyay S. Hydrophobically Modified Abelmoschus esculentus Polysaccharide Based Nanoparticles and Applications: A Review. Curr Drug Discov Technol 2022; 19:e010822207168. [PMID: 35927911 DOI: 10.2174/1570163819666220801121857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 04/07/2022] [Accepted: 05/13/2022] [Indexed: 01/27/2023]
Abstract
Nanomaterials are indeed a nanoscale technology that deals with the creation, evaluation, fabrication, and utilization of systems at the nanometre scale by manipulating their size and shape. We consider natural polysaccharides such as promising polysaccharides, which are biodegradable, nontoxic, abundant, and inexpensive bio-polymeric precursors for preparing the materials of choice in various industries. The aim is to review different methods to produce hydrophobically modified Abelmoschus esculentus nanoparticles and study the evaluation processes of these nanoparticles as given in the literature. It proved the benefits of derivatives of gum by introducing different chemical groups. The chemical functionalization of gum mainly includes the esterification, etherification, and crosslinking reactions of the hydroxyl groups and contains a special fibre which takes sugar levels in the blood under control, providing a sugar quantity suitable for the bowels. Okra contains mucilage that helps remove poisonous chemicals and bad cholesterol, often overloads the liver. Recovering from psychological conditions, like depression, general weakness, and joint healthiness can be done with Okra. Someone additionally applied it for pulmonary inflammation, bowel irritation, and sore throat. Purgative properties okra possesses are beneficial for bowel purification. It is used to counteract the acids. Fibre okra contains a valuable nutrient for intestinal microorganisms and ensures proper intestine functionality. It also protects the mucosa of the digestive tract by covering them with an extra layer because of its alkaline nature. Nanotechnology has emerged as a critical component of pharmaceutics, with many applications in drug carriers of interest aimed at improving drug clinical outcomes such as cancer, diabetes mellitus, wound care management, atopic dermatitis, cosmeceutical, etc. Beneficial outcomes of this review are discussed briefly.
Collapse
Affiliation(s)
- Prashant Upadhyay
- Faculty of Pharmacy, School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh, India
| | - Shivani Agarwal
- Faculty of Pharmacy, School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh, India
| | - Sukirti Upadhyay
- Faculty of Pharmacy, School of Pharmaceutical Sciences, IFTM University, Moradabad, Uttar Pradesh, India
| |
Collapse
|
24
|
Balakrishnan P, Gopi S. Highly efficient microencapsulation of phytonutrients by fractioned cellulose using biopolymer complexation technology. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2022; 19:607-618. [PMID: 35770826 DOI: 10.1515/jcim-2022-0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/29/2022] [Indexed: 06/15/2023]
Abstract
A poorly water soluble polar and non-polar bioactive complexes encapsulated in a nanocellulose-based polymeric network are the focus of this research. Ascorbic acid, resveratrol, holy basil extract, pomegranate extract, and niacin are all microencapsulated bioactive complexes that make up Zetalife®, a nutritional ingredient. It uses an interpenetrating polymeric network (IPN) with more dispersed nanocellulose and phospholipids to increase Zetalife® s bioavailability. Field Emission Scanning Electron Microscopic (FESEM) images were used in studying the morphology of encapsulated bioactive molecules. The average microbead size was determined to be 244.2 nm. After each month of storage, the sample's microbial content was measured to assess stability. In vitro release followed a first-order kinetic model with high R2.
Collapse
Affiliation(s)
- Preetha Balakrishnan
- Centre for Innovations and Technologies (CIT), ADSO Naturals Private Limited, Bangalore, India
- Research and Development, Curesupport B.V, Deventer, The Netherlands
| | - Sreerag Gopi
- Centre for Innovations and Technologies (CIT), ADSO Naturals Private Limited, Bangalore, India
- Research and Development, Curesupport B.V, Deventer, The Netherlands
| |
Collapse
|
25
|
Joun I, Nixdorf S, Deng W. Advances in lipid-based nanocarriers for breast cancer metastasis treatment. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:893056. [PMID: 36062261 PMCID: PMC9433809 DOI: 10.3389/fmedt.2022.893056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022] Open
Abstract
Breast cancer (BC) is the most common cancer affecting women worldwide, with over 2 million women diagnosed every year, and close to 8 million women currently alive following a diagnosis of BC in the last 5-years. The side effects such as chemodrug toxicity to healthy tissues and drug resistance severely affect the quality of life of BC patients. To overcome these limitations, many efforts have been made to develop nanomaterial-based drug delivery systems. Among these nanocarriers, lipid-based delivery platforms represented one of the most successful candidates for cancer therapy, improving the safety profile and therapeutic efficacy of encapsulated drugs. In this review we will mainly discuss and summarize the recent advances in such delivery systems for BC metastasis treatment, with a particular focus on targeting the common metastatic sites in bone, brain and lung. We will also provide our perspectives on lipid-based nanocarrier development for future clinical translation.
Collapse
Affiliation(s)
- Ingrid Joun
- School of Chemical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, Australia
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Sheri Nixdorf
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
| | - Wei Deng
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
- *Correspondence: Wei Deng
| |
Collapse
|
26
|
Cui J, Li W, Bu W, Liu J, Chen X, Li X, Liu C, Meng L, Chen M, Sun H, Wang J. Folic acid-modified disulfiram/Zn-IRMOF3 nanoparticles for oral cancer therapy by inhibiting ALDH1A1+ cancer stem cells. BIOMATERIALS ADVANCES 2022; 139:213038. [PMID: 35908474 DOI: 10.1016/j.bioadv.2022.213038] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/15/2022] [Accepted: 07/17/2022] [Indexed: 06/15/2023]
Abstract
The repurposing of old drugs can reduce the cost of drug development and speed up the availability of drugs for clinical use. Disulfiram (DSF) is an approved drug for alcohol abuse. In recent years, it has been established that DSF exerts an antitumor effect via targeted inhibition of ALDH1+ cancer stem cells (CSCs). However, due to its metal ion dependence, easy hydrolysis and low availability, the clinical application of DSF is limited. Previous studies have also shown that Zn2+ can inhibit CSCs. Accordingly, we developed a novel metal organic framework (IRMOF3)-Zn2+, and DSF was incorporated in the IRMOF3. Folic acid (FA) was subsequently loaded on the surface yielding IRMOF3 (IRMOF3-DSF-FA) for targeted therapy of tumors. The nanoscale IRMOF3-DSF-FA exhibited a high loading capacity, good biocompatibility and strong cell uptake capacity, which could provide metal ions, target tumor tissues and inhibit ALDH1+ CSCs. In vivo experiments showed that IRMOF3-DSF-FA could significantly inhibit the growth of CSCs and tumors, with no significant vital organ damage during treatment. Accordingly, IRMOF3-DSF-FA has great prospects for application as a DSF carrier, opening new horizons for targeted therapy of oral cancer.
Collapse
Affiliation(s)
- Jiasen Cui
- School and Hospital of Stomatology, Department of Oral Pathology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Weitao Li
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, P.O. Box 332, Shenyang 110819, China; College of Chemistry and Chemical Engineering, Xingtai University, Xingtai, Hebei 054001, China
| | - Wenhuan Bu
- School and Hospital of Stomatology, Department of Dental Materials, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Jinhui Liu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, P.O. Box 332, Shenyang 110819, China
| | - Xi Chen
- School and Hospital of Stomatology, Department of Oral Pathology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Xuewen Li
- School and Hospital of Stomatology, Department of Oral Pathology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Chunran Liu
- School and Hospital of Stomatology, Department of Oral Pathology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Lin Meng
- Department of Oral Pathology, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Mingli Chen
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, P.O. Box 332, Shenyang 110819, China.
| | - Hongchen Sun
- School and Hospital of Stomatology, Department of Oral Pathology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China.
| | - Jianhua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, P.O. Box 332, Shenyang 110819, China
| |
Collapse
|
27
|
Wang S, Cheng K, Chen K, Xu C, Ma P, Dang G, Yang Y, Lei Q, Huang H, Yu Y, Fang Y, Tang Q, Jiang N, Miao H, Liu F, Zhao X, Li N. Nanoparticle-based medicines in clinical cancer therapy. NANO TODAY 2022; 45:101512. [DOI: 10.1016/j.nantod.2022.101512] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
|
28
|
Qayyum S, Jabeen A, Aslam Z, Kanwal T, Shah MR, Faizi S. Synthesis and Characterization of Novel Lecithin Derived Nano-Formulation of Octyl and Dodecyl Gallate for Targeting B Cell Associated Non-Hodgkin’s Lymphoma. J CLUST SCI 2022. [DOI: 10.1007/s10876-022-02302-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Diego-González L, Fernández-Carrera A, Igea A, Martínez-Pérez A, Real Oliveira MECD, Gomes AC, Guerra C, Barbacid M, González-Fernández Á, Simón-Vázquez R. Combined Inhibition of FOSL-1 and YAP Using siRNA-Lipoplexes Reduces the Growth of Pancreatic Tumor. Cancers (Basel) 2022; 14:3102. [PMID: 35804874 PMCID: PMC9265026 DOI: 10.3390/cancers14133102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic cancer evades most of the current therapies and there is an urgent need for new treatments that could efficiently eliminate this aggressive tumor, such as the blocking of routes driving cell proliferation. In this work, we propose the use of small interfering RNA (siRNA) to inhibit the combined expression of FOSL-1 and YAP, two signaling proteins related with tumor cell proliferation and survival. To improve the efficacy of cell transfection, DODAB:MO (1:2) liposomes were used as siRNA nanocarriers, forming a complex denominated siRNA-lipoplexes. Liposomes and lipoplexes (carrying two siRNA for each targeted protein, or the combination of four siRNAs) were physico-chemically and biologically characterized. They showed very good biocompatibility and stability. The efficient targeting of FOSL-1 and YAP expression at both mRNA and protein levels was first proved in vitro using mouse pancreatic tumoral cell lines (KRASG12V and p53 knockout), followed by in vivo studies using subcutaneous allografts on mice. The peri-tumoral injection of lipoplexes lead to a significant decrease in the tumor growth in both Athymic Nude-Foxn1nu and C57BL/6 mice, mainly in those receiving the combination of four siRNAs, targeting both YAP and FOSL-1. These results open a new perspective to overcome the fast tumor progression in pancreatic cancer.
Collapse
Affiliation(s)
- Lara Diego-González
- CINBIO, Universidade de Vigo, Immunology Group, 36310 Vigo, Spain; (L.D.-G.); (A.F.-C.); (A.I.); (A.M.-P.); (Á.G.-F.)
- Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain
| | - Andrea Fernández-Carrera
- CINBIO, Universidade de Vigo, Immunology Group, 36310 Vigo, Spain; (L.D.-G.); (A.F.-C.); (A.I.); (A.M.-P.); (Á.G.-F.)
- Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain
| | - Ana Igea
- CINBIO, Universidade de Vigo, Immunology Group, 36310 Vigo, Spain; (L.D.-G.); (A.F.-C.); (A.I.); (A.M.-P.); (Á.G.-F.)
- Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain
| | - Amparo Martínez-Pérez
- CINBIO, Universidade de Vigo, Immunology Group, 36310 Vigo, Spain; (L.D.-G.); (A.F.-C.); (A.I.); (A.M.-P.); (Á.G.-F.)
- Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain
| | | | - Andreia C. Gomes
- CBMA (Centre of Molecular and Environmental Biology), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal;
| | - Carmen Guerra
- CNIO (Centro Nacional de Investigaciones Oncológicas), Experimental Oncology Group, 28029 Madrid, Spain; (C.G.); (M.B.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mariano Barbacid
- CNIO (Centro Nacional de Investigaciones Oncológicas), Experimental Oncology Group, 28029 Madrid, Spain; (C.G.); (M.B.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - África González-Fernández
- CINBIO, Universidade de Vigo, Immunology Group, 36310 Vigo, Spain; (L.D.-G.); (A.F.-C.); (A.I.); (A.M.-P.); (Á.G.-F.)
- Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain
| | - Rosana Simón-Vázquez
- CINBIO, Universidade de Vigo, Immunology Group, 36310 Vigo, Spain; (L.D.-G.); (A.F.-C.); (A.I.); (A.M.-P.); (Á.G.-F.)
- Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), SERGAS-UVIGO, 36312 Vigo, Spain
| |
Collapse
|
30
|
Mojarad-Jabali S, Mahdinloo S, Farshbaf M, Sarfraz M, Fatahi Y, Atyabi F, Valizadeh H. Transferrin receptor-mediated liposomal drug delivery: recent trends in targeted therapy of cancer. Expert Opin Drug Deliv 2022; 19:685-705. [PMID: 35698794 DOI: 10.1080/17425247.2022.2083106] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Compared to normal cells, malignant cancer cells require more iron for their growth and rapid proliferation, which can be supplied by a high expression level of transferrin receptor (TfR). It is well known that the expression of TfR on the tumor cells is considerably higher than that of normal cells, which makes TfR an attractive target in cancer therapy. AREAS COVERED In this review, the primary focus is on the role of TfR as a valuable tool for cancer-targeted drug delivery, followed by the full coverage of available TfR ligands and their conjugation chemistry to the surface of liposomes. Finally, the most recent studies investigating the potential of TfR-targeted liposomes as promising drug delivery vehicles to different cancer cells are highlighted with emphasis on their improvement possibilities to become a part of future cancer medicines. EXPERT OPINION Liposomes as a valuable class of nanocarriers have gained much attention toward cancer therapy. From all the studies that have exploited the therapeutic and diagnostic potential of TfR on cancer cells, it can be realized that the systematic assessment of TfR ligands applied for liposomal targeted delivery has yet to be entirely accomplished.
Collapse
Affiliation(s)
- Solmaz Mojarad-Jabali
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Student research committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Mahdinloo
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Student research committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Farshbaf
- Student research committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Muhammad Sarfraz
- College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates
| | - Yousef Fatahi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hadi Valizadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
31
|
Xu Y, Fourniols T, Labrak Y, Préat V, Beloqui A, des Rieux A. Surface Modification of Lipid-Based Nanoparticles. ACS NANO 2022; 16:7168-7196. [PMID: 35446546 DOI: 10.1021/acsnano.2c02347] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
There is a growing interest in the development of lipid-based nanocarriers for multiple purposes, including the recent increase of these nanocarriers as vaccine components during the COVID-19 pandemic. The number of studies that involve the surface modification of nanocarriers to improve their performance (increase the delivery of a therapeutic to its target site with less off-site accumulation) is enormous. The present review aims to provide an overview of various methods associated with lipid nanoparticle grafting, including techniques used to separate grafted nanoparticles from unbound ligands or to characterize grafted nanoparticles. We also provide a critical perspective on the usefulness and true impact of these modifications on overcoming different biological barriers, with our prediction on what to expect in the near future in this field.
Collapse
Affiliation(s)
- Yining Xu
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Thibaut Fourniols
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Yasmine Labrak
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 72 B1.72.01, 1200 Brussels, Belgium
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Ana Beloqui
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Anne des Rieux
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| |
Collapse
|
32
|
dos Santos AM, Junior AGT, Carvalho SG, Chorilli M. An updated review on properties, nanodelivery systems, and analytical methods for the determination of 5-fluorouracil in pharmaceutical and biological samples. Curr Pharm Des 2022; 28:1501-1512. [DOI: 10.2174/1381612828666220509150918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 03/22/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
5-Fluorouracil (5-FU) is an antimetabolite drug used for over 70 years as first-line chemotherapy to treat various types of cancer, such as head, neck, breast and colorectal cancer. 5-FU acts mainly by inhibiting thymidylate synthase, thereby interfering with deoxyribonucleic acid (DNA) replication or by 5-FU incorporating into DNA, causing damage to the sequence of nucleotides. Being analogous to uracil, 5-FU enters cells using the same transport mechanism, where a is converted into active metabolites such as fluorouridine triphosphate (FUTP), fluorodeoxyuridine monophosphate (FdUMP), and fluorodeoxyuridine triphosphate (FdUTP). Currently, there are several nanodelivery systems being developed and evaluated at the preclinical level to overcome existing limitations to 5-FU chemotherapy, including liposomes, polymeric nanoparticles, polymeric micelles, nanoemulsions, mesoporous silica nanoparticles, and solid lipid nanoparticles. Therefore, it is essential to choose and develop suitable analytical methods for the quantification of 5-FU and its metabolites (5-fluorouridine and 5-fluoro-2-deoxyuridine) in pharmaceutical and biological samples. Among the analytical techniques, chromatographic methods are commonly the most used for the quantification of 5-FU from different matrices. However, other analytical methods have also been developed for the determination of 5-FU, such as electrochemical methods, a sensitive, selective, and precise technique, in addition to having a reduced cost. Here, we first review the physicochemical properties, mechanism of action, and advances in 5-FU nanodelivery systems. Next, we summarize the current progress of other chromatographic methods described to determine 5-FU. Lastly, we discuss the advantages of electrochemical methods for the identification and quantification of 5-FU and its metabolites in pharmaceutical and biological samples.
Collapse
Affiliation(s)
- Aline Martins dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, 14800-903, Brazil
| | | | - Suzana Gonçalves Carvalho
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, 14800-903, Brazil
| | - Marlus Chorilli
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, SP, 14800-903, Brazil
| |
Collapse
|
33
|
Nanoencapsulation of aptamer-functionalized 5-Fluorouracil liposomes using alginate/chitosan complex as a novel targeting strategy for colon-specific drug delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
34
|
Mechanisms of Penetration Enhancement and Transport Utilizing Skin Keratine Liposomes for the Topical Delivery of Licochalcone A. Molecules 2022; 27:molecules27082504. [PMID: 35458701 PMCID: PMC9029797 DOI: 10.3390/molecules27082504] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Keratin liposomes have emerged as a useful topical drug delivery system given theirenhanced ability to penetrate the skin, making them ideal as topical drug vehicles. However, the mechanisms of the drug penetration enhancement of keratin liposomes have not been clearly elucidated. Therefore, licochalcone A(LA)-loaded skin keratin liposomes (LALs) were prepared to investigate their mechanisms of penetration enhancement on the skin and inB16F10 cells. Skin deposition studies, differential scanning calorimetry (DSC), attenuated total reflection-Fourier Transform Infrared Spectroscopy (ATR-FTIR), and skin distribution and intracellular distribution studies were carried out to demonstrate the drug enhancement mechanisms of LALs. We found that the optimal application of LALs enhanced drug permeation via alterations in the components, structure, and thermodynamic properties of the stratum corneum (SC), that is, by enhancing the lipid fluidization, altering the skin keratin, and changing the thermodynamic properties of the SC. Moreover, hair follicles were the main penetration pathways for the LA delivery, which occurred in a time-dependent manner. In the B16F10 cells, the skin keratin liposomes effectively delivered LA into the cytoplasm without cytotoxicity. Thus, LAL nanoparticles are promising topical drug delivery systems for pharmaceutical and cosmetic applications.
Collapse
|
35
|
Liu P, Chen G, Zhang J. A Review of Liposomes as a Drug Delivery System: Current Status of Approved Products, Regulatory Environments, and Future Perspectives. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041372. [PMID: 35209162 PMCID: PMC8879473 DOI: 10.3390/molecules27041372] [Citation(s) in RCA: 361] [Impact Index Per Article: 120.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/03/2022] [Accepted: 02/13/2022] [Indexed: 12/12/2022]
Abstract
Liposomes have been considered promising and versatile drug vesicles. Compared with traditional drug delivery systems, liposomes exhibit better properties, including site-targeting, sustained or controlled release, protection of drugs from degradation and clearance, superior therapeutic effects, and lower toxic side effects. Given these merits, several liposomal drug products have been successfully approved and used in clinics over the last couple of decades. In this review, the liposomal drug products approved by the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA) are discussed. Based on the published approval package in the FDA and European public assessment report (EPAR) in EMA, the critical chemistry information and mature pharmaceutical technologies applied in the marketed liposomal products, including the lipid excipient, manufacturing methods, nanosizing technique, drug loading methods, as well as critical quality attributions (CQAs) of products, are introduced. Additionally, the current regulatory guidance and future perspectives related to liposomal products are summarized. This knowledge can be used for research and development of the liposomal drug candidates under various pipelines, including the laboratory bench, pilot plant, and commercial manufacturing.
Collapse
Affiliation(s)
- Peng Liu
- Correspondence: (P.L.); (J.Z.); Tel.: +86-1332-1952-664 (P.L.); +86-1891-7601-368 (J.Z.)
| | | | - Jingchen Zhang
- Correspondence: (P.L.); (J.Z.); Tel.: +86-1332-1952-664 (P.L.); +86-1891-7601-368 (J.Z.)
| |
Collapse
|
36
|
Storage Stability and In Vitro Bioaccessibility of Liposomal Betacyanins from Red Pitaya ( Hylocereus polyrhizus). Molecules 2022; 27:molecules27041193. [PMID: 35208980 PMCID: PMC8880447 DOI: 10.3390/molecules27041193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/30/2022] [Accepted: 02/07/2022] [Indexed: 11/22/2022] Open
Abstract
In order to address the poor stability of the betacyanins from red pitaya (Hylocereus polyrhizus, HP), which are considered as good sources of natural colorant, liposomal-encapsulation technique was applied in this study. Thin-layer dispersion method was employed to prepare HP betacyacnin liposomes (HPBL). The formulation parameters for HPBL were optimized, and the characteristics, stability, and release profile of HPBL in in vitro gastrointestinal systems were evaluated.Results showed that an HP betacyanin encapsulation efficiency of 93.43 ± 0.11% was obtained after formulation optimization. The HPBL exhibited a narrow size distribution of particle within a nanometer range and a strong electronegative ζ-potential. By liposomal encapsulation, storage stability of HP betacyanin was significantly enhanced in different storage temperatures. When the environmental pH ranged from 4.3–7.0, around 80% of HP betacyanins were preserved on Day 21 with the liposomal protection. The loss of 2,2′-Diphenyl-picrylhydrazyl (DPPH) scavenging activity and color deterioration of HPBL were developed in accordance with the degradation of HP betacyanins during storage. In in vitro gastrointestinal digestion study, with the protection of liposome, the retention rates of HP betacyanins in vitro were enhanced by 14% and 40% for gastric and intestinal digestion, respectively.This study suggested that liposomal encapsulation was an effective approach to stabilize HP betacyanins during storage and gastrointestinal digestion, but further investigations were needed to better optimize the liposomal formulation and understand the complex liposomal system.
Collapse
|
37
|
Mirveis Z, Kouchak M, Mahdavinia M, Rahbar N. Novel and efficient method for loading aptamer-conjugated liposomes with arsenic trioxide for targeting cancer cells. J Liposome Res 2021; 32:276-283. [PMID: 34918592 DOI: 10.1080/08982104.2021.2005624] [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: 10/19/2022]
Abstract
Although the therapeutic effect of liposomal arsenic trioxide arsenic trioxide (ATO) in the treatment of solid tumours has been confirmed, its dose-limiting loading is a challenging issue. To solve the problems in the preparation of liposomal ATO, different loading strategies were evaluated and compared. In addition, liposomes decorated with anti-nucleolin aptamers were developed as a novel formulation for targeted delivery with high loading efficiency and sustained releasing property in order to treat solid tumours. The liposomes were prepared by a thin-film method exploiting the passive loading strategy of Co(II) hydrogen arsenite (CHA). The structural characteristics of the liposomes were also investigated by Fourier transform infra-red spectroscopy (FT-IR), dynamic light scattering (DLS), zeta potentiometry, field emission scanning electron microscopy (FESEM), and Energy Dispersive X-ray Diffraction (EDX) techniques. To evaluate the potential cytotoxicity of this liposomal drug vehicle in vitro, MTT assay was performed on HT-29 cancer cell line. The results showed that the synthesised liposomes loaded with CHA exhibited high entrapment efficiency (77%). MTT assays showed a significant difference between the percentage of viable cells when HT -29 cells were treated with free ATO and liposomal formulation which can be corresponded to the sustained release of the drug from the liposomes. The results of this study may lead to a promising strategy for the effective treatment of solid tumours.
Collapse
Affiliation(s)
- Zohreh Mirveis
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nadereh Rahbar
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
38
|
Zhang H, Gao Z, Li X, Li L, Ye S, Tang B. Multiple-mRNA-controlled and heat-driven drug release from gold nanocages in targeted chemo-photothermal therapy for tumors. Chem Sci 2021; 12:12429-12436. [PMID: 34603673 PMCID: PMC8480318 DOI: 10.1039/d1sc02017j] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/06/2021] [Indexed: 12/30/2022] Open
Abstract
Multifunctional drug delivery systems enabling effective drug delivery and comprehensive treatment are critical to successful cancer treatment. Overcoming nonspecific release and off-target effects remains challenging in precise drug delivery. Here, we design triple-interlocked drug delivery systems to perform specific cancer cell recognition, controlled drug release and effective comprehensive therapy. Gold nanocages (AuNCs) comprise a novel class of nanostructures possessing hollow interiors and porous walls. AuNCs are employed as a drug carrier and photothermal transducer due to their unique structure and photothermal properties. A smart triple-interlocked I-type DNA nanostructure is modified on the surface of the AuNCs, and molecules of the anticancer drug doxorubicin (DOX) are loaded as molecular cargo and blocked. The triple-interlocked nanostructure can be unlocked by binding with three types of tumor-related mRNAs, which act as "keys" to the triple locks, sequentially, which leads to precise drug release. Additionally, fluorescence-imaging-oriented chemical-photothermal synergistic treatment is achieved under illumination with infrared light. This drug delivery system, which combines the advantages of AuNCs and interlocked I-type DNA, successfully demonstrates effective and precise imaging, drug release and photothermal therapy. This multifunctional triple-interlocked drug delivery system could be used as a potential carrier for effective cancer-targeting comprehensive chemotherapy and photothermal therapy treatments.
Collapse
Affiliation(s)
- Hao Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University Jinan 250014 P. R. China
| | - Ze Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University Jinan 250014 P. R. China
| | - Xiaoxiao Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University Jinan 250014 P. R. China
| | - Lu Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University Jinan 250014 P. R. China
| | - Sujuan Ye
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, State Key Laboratory Base for Eco-chemical Engineering, College of Science and Technology Qingdao 266042 P. R.China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University Jinan 250014 P. R. China
| |
Collapse
|
39
|
Hani U, M. YB, Wahab S, Siddiqua A, Osmani RAM, Rahamathulla M. A Comprehensive Review of Current Perspectives on Novel Drug Delivery Systems and Approaches for Lung Cancer Management. J Pharm Innov 2021. [DOI: 10.1007/s12247-021-09582-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
40
|
Bartels K, Lasitza‐Male T, Hofmann H, Löw C. Single-Molecule FRET of Membrane Transport Proteins. Chembiochem 2021; 22:2657-2671. [PMID: 33945656 PMCID: PMC8453700 DOI: 10.1002/cbic.202100106] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/03/2021] [Indexed: 12/31/2022]
Abstract
Uncovering the structure and function of biomolecules is a fundamental goal in structural biology. Membrane-embedded transport proteins are ubiquitous in all kingdoms of life. Despite structural flexibility, their mechanisms are typically studied by ensemble biochemical methods or by static high-resolution structures, which complicate a detailed understanding of their dynamics. Here, we review the recent progress of single molecule Förster Resonance Energy Transfer (smFRET) in determining mechanisms and timescales of substrate transport across membranes. These studies do not only demonstrate the versatility and suitability of state-of-the-art smFRET tools for studying membrane transport proteins but they also highlight the importance of membrane mimicking environments in preserving the function of these proteins. The current achievements advance our understanding of transport mechanisms and have the potential to facilitate future progress in drug design.
Collapse
Affiliation(s)
- Kim Bartels
- Centre for Structural Systems Biology (CSSB)DESY and European Molecular Biology Laboratory HamburgNotkestrasse 8522607HamburgGermany
| | - Tanya Lasitza‐Male
- Department of Structural BiologyWeizmann Institute of ScienceHerzl St. 2347610001RehovotIsrael
| | - Hagen Hofmann
- Department of Structural BiologyWeizmann Institute of ScienceHerzl St. 2347610001RehovotIsrael
| | - Christian Löw
- Centre for Structural Systems Biology (CSSB)DESY and European Molecular Biology Laboratory HamburgNotkestrasse 8522607HamburgGermany
| |
Collapse
|
41
|
Park JY, Shin Y, Won WR, Lim C, Kim JC, Kang K, Husni P, Lee ES, Youn YS, Oh KT. Development of AE147 Peptide-Conjugated Nanocarriers for Targeting uPAR-Overexpressing Cancer Cells. Int J Nanomedicine 2021; 16:5437-5449. [PMID: 34408417 PMCID: PMC8367088 DOI: 10.2147/ijn.s315619] [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: 04/15/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose An AE147 peptide-conjugated nanocarrier based on PEGylated liposomes was developed in order to target the metastatic tumors overexpressing urokinase-type plasminogen activator receptor (uPAR), which cancer progression via uPA signaling. Therefore, the AE147 peptide-conjugated nanocarrier system may hold the potential for active targeting of metastatic tumors. Methods The AE147 peptide, an antagonist of uPAR, was conjugated to the PEGylated liposomes for targeting metastatic tumors overexpressing uPAR. Docetaxel (DTX), an anticancer drug, was incorporated into the nanocarriers. The structure of the AE147-conjugated nanocarrier, its physicochemical properties, and in vivo biodistribution were evaluated. Results The DTX-loaded nanocarrier showed a spherical structure, a high drug-loading capacity, and a high colloidal stability. Drug carrying AE147 conjugates were actively taken up by the uPAR-overexpressing MDA-MB-231 cancer cells. In vivo animal imaging confirmed that the AE147-conjugated nanoparticles effectively accumulated at the sites of tumor metastasis. Conclusion The AE147-nanocarrier showed potential for targeting metastatic tumor cells overexpressing uPAR and as a nanomedicine platform for theragnosis applications. These results suggest that this novel nano-platform will facilitate further advancements in cancer therapy.
Collapse
Affiliation(s)
- June Yong Park
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Yuseon Shin
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Woong Roeck Won
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Chaemin Lim
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea.,Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jae Chang Kim
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Kioh Kang
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Patihul Husni
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Eun Seong Lee
- Division of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Yu Seok Youn
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Kyung Taek Oh
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea.,College of Pharmacy, Chung-Ang University, Dongjak-gu, Seoul, 06974, Republic of Korea
| |
Collapse
|
42
|
Chuffa LGDA, Seiva FRF, Novais AA, Simão VA, Martín Giménez VM, Manucha W, Zuccari DAPDC, Reiter RJ. Melatonin-Loaded Nanocarriers: New Horizons for Therapeutic Applications. Molecules 2021; 26:molecules26123562. [PMID: 34200947 PMCID: PMC8230720 DOI: 10.3390/molecules26123562] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
The use of nanosized particles has emerged to facilitate selective applications in medicine. Drug-delivery systems represent novel opportunities to provide stricter, focused, and fine-tuned therapy, enhancing the therapeutic efficacy of chemical agents at the molecular level while reducing their toxic effects. Melatonin (N-acetyl-5-methoxytriptamine) is a small indoleamine secreted essentially by the pineal gland during darkness, but also produced by most cells in a non-circadian manner from which it is not released into the blood. Although the therapeutic promise of melatonin is indisputable, aspects regarding optimal dosage, biotransformation and metabolism, route and time of administration, and targeted therapy remain to be examined for proper treatment results. Recently, prolonged release of melatonin has shown greater efficacy and safety when combined with a nanostructured formulation. This review summarizes the role of melatonin incorporated into different nanocarriers (e.g., lipid-based vesicles, polymeric vesicles, non-ionic surfactant-based vesicles, charge carriers in graphene, electro spun nanofibers, silica-based carriers, metallic and non-metallic nanocomposites) as drug delivery system platforms or multilevel determinations in various in vivo and in vitro experimental conditions. Melatonin incorporated into nanosized materials exhibits superior effectiveness in multiple diseases and pathological processes than does free melatonin; thus, such information has functional significance for clinical intervention.
Collapse
Affiliation(s)
- Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP-São Paulo State University, Botucatu, São Paulo 18618-689, Brazil; (L.G.d.A.C.); (V.A.S.)
| | - Fábio Rodrigues Ferreira Seiva
- Biological Science Center, Department of Biology, Luiz Meneghel Campus, Universidade Estadual do Norte do Paraná-UENP, Bandeirantes 86360-000, PR, Brazil;
| | - Adriana Alonso Novais
- Health Sciences Institute, Federal University of Mato Grosso, UFMT, Sinop 78607-059, MG, Brazil;
| | - Vinícius Augusto Simão
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP-São Paulo State University, Botucatu, São Paulo 18618-689, Brazil; (L.G.d.A.C.); (V.A.S.)
| | - Virna Margarita Martín Giménez
- Facultad de Ciencias Químicas y Tecnológicas, Instituto de Investigaciones en Ciencias Químicas, Universidad Católica de Cuyo, Sede San Juan 5400, Argentina;
| | - Walter Manucha
- Laboratorio de Farmacología Experimental Básica y Traslacional. Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina;
- Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigación Científica y Tecnológica (IMBECU-CONICET), Mendoza 5500, Argentina
| | | | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA
- Correspondence:
| |
Collapse
|
43
|
Sang R, Stratton B, Engel A, Deng W. Liposome technologies towards colorectal cancer therapeutics. Acta Biomater 2021; 127:24-40. [PMID: 33812076 DOI: 10.1016/j.actbio.2021.03.055] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/05/2021] [Accepted: 03/23/2021] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the fourth most common deadly cancer worldwide. After treatment with curative intent recurrence rates vary with staging 0-13% in Stage 1, 11-61% in S2 and 28-73% in Stage 3. The toxicity to healthy tissues from chemotherapy and radiotherapy and drug resistance severely affect the quality of life and cancer specific outcomes of CRC patients. To overcome some of these limitations, many efforts have been made to develop nanomaterial-based drug delivery systems. Among these nanocarriers, liposomes represented one of the most successful candidates in delivering targeted oncological treatment, improving safety profile and therapeutic efficacy of encapsulated drugs. In this review we will discuss liposome design with a particular focus on the targeting feature and triggering functions. We will also summarise the recent advances in liposomal delivery system for CRC treatment in both the preclinical and clinical studies. We will finally provide our perspectives on the liposome technology development for the future clinical translation. STATEMENT OF SIGNIFICANCE: Conventional treatments for colorectal cancer (CRC) severely affect the therapeutic effects for advanced patients. With the development of nanomedicines, liposomal delivery system appears to be one of the most promising nanocarriers for CRC treatment. In last three years several reviews in this area have been published focusing on the preclinical research and drug delivery function, which is a fairly narrow focus in the field of liposome technology for CRC therapy. Our review presented the most recent advances of the liposome technology (both clinical and preclinical applications) for CRC with strong potential for further clinical translation. We believe it will attract lots of attention from various audiences, including researchers, clinicians and the industry.
Collapse
|
44
|
Mojarad-Jabali S, Farshbaf M, Walker PR, Hemmati S, Fatahi Y, Zakeri-Milani P, Sarfraz M, Valizadeh H. An update on actively targeted liposomes in advanced drug delivery to glioma. Int J Pharm 2021; 602:120645. [PMID: 33915182 DOI: 10.1016/j.ijpharm.2021.120645] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022]
Abstract
High-grade glioma is one of the most aggressive types of cancer with a low survival rate ranging from 12 to 15 months after the first diagnosis. Though being the most common strategy for glioma therapy, conventional chemotherapy suffers providing the therapeutic dosage of common therapeutics mostly because of limited permeability of blood-brain barrier (BBB), and blood-brain tumor barrier (BBTB) to anticancer agents. Among various nanoformulations, liposomes are considered as the most popular carriers aimed for glioma therapy. However, non-targeted liposomes which passively accumulate in most of the cancer tissues mainly through the enhanced permeation and retention effect (EPR), may not be applicable for glioma therapy due to BBB tight junctions. In the recent decade, the surface modification of liposomes with different active targeting ligands has shown promising results by getting different chemotherapeutics across the BBB and BBTB and leading them into the glioma cells. The present review discusses the major barriers for drug delivery systems to glioma, elaborates the existing mechanisms for liposomes to traverse across the BBB, and explores the main strategies for incorporation of targeting ligands onto the liposomes. It subsequently investigates the most recent and relevant studies of actively targeted liposomes modified with antibodies, aptamers, monosaccharides, polysaccharides, proteins, and peptides applied for effective glioma therapy, and highlights the common challenges facing this area. Finally, the actively targeted liposomes undergoing preclinical and clinical studies for delivery of different anticancer agents to glioma cells will be reviewed.
Collapse
Affiliation(s)
- Solmaz Mojarad-Jabali
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Farshbaf
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Paul R Walker
- Center for Translational Research in Onco-Hematology, Department of Medicine, University of Geneva and Division of Oncology, Geneva University Hospitals, Geneva, Switzerland
| | - Salar Hemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Fatahi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Muhammad Sarfraz
- College of Pharmacy, Al Ain University, Al Ain 64141, United Arab Emirates
| | - Hadi Valizadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
45
|
Lipid nanovesicles for biomedical applications: 'What is in a name'? Prog Lipid Res 2021; 82:101096. [PMID: 33831455 DOI: 10.1016/j.plipres.2021.101096] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/28/2021] [Accepted: 03/28/2021] [Indexed: 12/12/2022]
Abstract
Vesicles, generally defined as self-assembled structures formed by single or multiple concentric bilayers that surround an aqueous core, have been widely used for biomedical applications. They can either occur naturally (e.g. exosomes) or be produced artificially and range from the micrometric scale to the nanoscale. One the most well-known vesicle is the liposome, largely employed as a drug delivery nanocarrier. Liposomes have been modified along the years to improve physicochemical and biological features, resulting in long-circulating, ligand-targeted and stimuli-responsive liposomes, among others. In this process, new nomenclatures were reported in an extensive literature. In many instances, the new names suggest the emergence of a new nanocarrier, which have caused confusion as to whether the vesicles are indeed new entities or could simply be considered modified liposomes. Herein, we discussed the extensive nomenclature of vesicles based on the suffix "some" that are employed for drug delivery and composed of various types and proportions of lipids and others amphiphilic compounds. New names have most often been selected based on changes of vesicle lipid composition, but the payload, structural complexity (e.g. multicompartment) and new/improved proprieties (e.g. elasticity) have also inspired new vesicle names. Based on this discussion, we suggested a rational classification for vesicles.
Collapse
|
46
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
47
|
Ferreira M, Ogren M, Dias JNR, Silva M, Gil S, Tavares L, Aires-da-Silva F, Gaspar MM, Aguiar SI. Liposomes as Antibiotic Delivery Systems: A Promising Nanotechnological Strategy against Antimicrobial Resistance. Molecules 2021; 26:2047. [PMID: 33918529 PMCID: PMC8038399 DOI: 10.3390/molecules26072047] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022] Open
Abstract
Antimicrobial drugs are key tools to prevent and treat bacterial infections. Despite the early success of antibiotics, the current treatment of bacterial infections faces serious challenges due to the emergence and spread of resistant bacteria. Moreover, the decline of research and private investment in new antibiotics further aggravates this antibiotic crisis era. Overcoming the complexity of antimicrobial resistance must go beyond the search of new classes of antibiotics and include the development of alternative solutions. The evolution of nanomedicine has allowed the design of new drug delivery systems with improved therapeutic index for the incorporated compounds. One of the most promising strategies is their association to lipid-based delivery (nano)systems. A drug's encapsulation in liposomes has been demonstrated to increase its accumulation at the infection site, minimizing drug toxicity and protecting the antibiotic from peripheral degradation. In addition, liposomes may be designed to fuse with bacterial cells, holding the potential to overcome antimicrobial resistance and biofilm formation and constituting a promising solution for the treatment of potential fatal multidrug-resistant bacterial infections, such as methicillin resistant Staphylococcus aureus. In this review, we aim to address the applicability of antibiotic encapsulated liposomes as an effective therapeutic strategy for bacterial infections.
Collapse
Affiliation(s)
- Magda Ferreira
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria Ogren
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Joana N. R. Dias
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Marta Silva
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Solange Gil
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Luís Tavares
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Frederico Aires-da-Silva
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Sandra Isabel Aguiar
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| |
Collapse
|
48
|
Naik H, Sonju JJ, Singh S, Chatzistamou I, Shrestha L, Gauthier T, Jois S. Lipidated Peptidomimetic Ligand-Functionalized HER2 Targeted Liposome as Nano-Carrier Designed for Doxorubicin Delivery in Cancer Therapy. Pharmaceuticals (Basel) 2021; 14:221. [PMID: 33800723 PMCID: PMC8002094 DOI: 10.3390/ph14030221] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
The therapeutic index of chemotherapeutic agents can be improved by the use of nano-carrier-mediated chemotherapeutic delivery. Ligand-targeted drug delivery can be used to achieve selective and specific delivery of chemotherapeutic agents to cancer cells. In this study, we prepared a peptidomimetic conjugate (SA-5)-tagged doxorubicin (Dox) incorporated liposome (LP) formulation (SA-5-Dox-LP) to evaluate the targeted delivery potential of SA-5 in human epidermal growth factor receptor-2 (HER2) overexpressed non-small-cell lung cancer (NSCLC) and breast cancer cell lines. The liposome was prepared using thin lipid film hydration and was characterized for particle size, encapsulation efficiency, cell viability, and targeted cellular uptake. In vivo evaluation of the liposomal formulation was performed in a mice model of NSCLC. The cell viability studies revealed that targeted SA-5-Dox-LP showed better antiproliferative activity than non-targeted Dox liposomes (Dox-LP). HER2-targeted liposome delivery showed selective cellular uptake compared to non-targeted liposomes on cancer cells. In vitro drug release studies indicated that Dox was released slowly from the formulations over 24 h, and there was no difference in Dox release between Dox-LP formulation and SA-5-Dox-LP formulation. In vivo studies in an NSCLC model of mice indicated that SA-5-Dox-LP could reduce the lung tumors significantly compared to vehicle control and Dox. In conclusion, this study demonstrated that the SA-5-Dox-LP liposome has the potential to increase therapeutic efficiency and targeted delivery of Dox in HER2 overexpressing cancer.
Collapse
Affiliation(s)
- Himgauri Naik
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA; (H.N.); (J.J.S.); (S.S.); (L.S.)
| | - Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA; (H.N.); (J.J.S.); (S.S.); (L.S.)
| | - Sitanshu Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA; (H.N.); (J.J.S.); (S.S.); (L.S.)
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology & Immunology (PMI), School of Medicine, USC, SC 6439 Garners Ferry Rd, Columbia, SC 29208, USA;
| | - Leeza Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA; (H.N.); (J.J.S.); (S.S.); (L.S.)
| | - Ted Gauthier
- Biotechnology Laboratory, LSU AgCenter, Louisiana State University, Baton Rouge, LA 70803, USA;
| | - Seetharama Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA; (H.N.); (J.J.S.); (S.S.); (L.S.)
| |
Collapse
|
49
|
Meng W, He C, Hao Y, Wang L, Li L, Zhu G. Prospects and challenges of extracellular vesicle-based drug delivery system: considering cell source. Drug Deliv 2021; 27:585-598. [PMID: 32264719 PMCID: PMC7178886 DOI: 10.1080/10717544.2020.1748758] [Citation(s) in RCA: 335] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, are nanosized membrane vesicles derived from most cell types. Carrying diverse biomolecules from their parent cells, EVs are important mediators of intercellular communication and thus play significant roles in physiological and pathological processes. Owing to their natural biogenesis process, EVs are generated with high biocompatibility, enhanced stability, and limited immunogenicity, which provide multiple advantages as drug delivery systems (DDSs) over traditional synthetic delivery vehicles. EVs have been reported to be used for the delivery of siRNAs, miRNAs, protein, small molecule drugs, nanoparticles, and CRISPR/Cas9 in the treatment of various diseases. As a natural drug delivery vectors, EVs can penetrate into the tissues and be bioengineered to enhance the targetability. Although EVs' characteristics make them ideal for drug delivery, EV-based drug delivery remains challenging, due to lack of standardized isolation and purification methods, limited drug loading efficiency, and insufficient clinical grade production. In this review, we summarized the current knowledge on the application of EVs as DDS from the perspective of different cell origin and weighted the advantages and bottlenecks of EV-based DDS.
Collapse
Affiliation(s)
- Wanrong Meng
- Department of Stomatology, School of Medicine, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Chanshi He
- Department of Stomatology, School of Medicine, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Yaying Hao
- Department of Stomatology, School of Medicine, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Linlin Wang
- Department of Stomatology, School of Medicine, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Ling Li
- Department of Stomatology, School of Medicine, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Guiquan Zhu
- Department of Stomatology, School of Medicine, Sichuan Cancer Hospital, Sichuan Key Laboratory of Radiation Oncology, University of Electronic Science and Technology of China, Chengdu, PR China
| |
Collapse
|
50
|
|