1
|
Hamze Mostafavi S, Mohammadi S, Mousavi Alborzi FS, Hajiahmadi F, Ahmadvand D, Gheibi N, Naderi-Manesh H, Shariatifar H, Farasat A. TRA/MEL immunoliposomes act as a targeted medicine in BT-474 breast cancer cells. J Liposome Res 2025:1-11. [PMID: 40396920 DOI: 10.1080/08982104.2025.2505102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 04/23/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
Breast cancer is one of the most common and deadly cancers worldwide. Melittin is the main component of bee venom, which has multiple anti-cancer properties. Targeted delivery of the gene encoding melittin using TRA-conjugated immunoliposomes to breast cancer cells can effectively treat this disease and reduce the side effects. Liposomes were prepared using the thin-film hydration method. The conjugation of TRA to liposomes was confirmed using SDS-PAGE, FTIR, and Bradford assay and characterized by DLS and TEM. The MTT, Fluorescent microscopy imaging, and flow cytometry methods were chosen to investigate the cytotoxicity and internalization of MEL/PEG-Lip and TRA/MEL immunoliposomes in the BT-474 cell line. The hydrodynamic diameter of TRA/MEL immunoliposomes was about 156 nm, and their appearance was spherical. The IC50 values for TRA/MEL immunoliposomes were calculated as 7.73 and 5.41 µg/mL for 48 and 72 h, respectively, which indicated that TRA/MEL immunoliposomes had a more significant cytotoxic effect on BT-474 cells than MEL/PEG-Lip. In addition, flow cytometry results showed that TRA/MEL immunoliposomes enter BT-474 cells to a greater extent and cause apoptosis. Due to the ability of TRA/MEL immunoliposomes to target and induce apoptosis in BT-474 cancer cells, this nanostructure can be suggested as a promising alternative in the treatment of this type of breast cancer.
Collapse
Affiliation(s)
- Sajjad Hamze Mostafavi
- Student Research Committee, School of Paramedical, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sahar Mohammadi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Fahimeh Hajiahmadi
- Cellular Molecular Pharmacology School, School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Davoud Ahmadvand
- Department of Medical Imaging Technology and Molecular Imaging, Faculty of advanced technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hossein Naderi-Manesh
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hanifeh Shariatifar
- Food and Drug Laboratory Research Center, Food and Drug Administration, MOH & ME, Tehran, Iran
| | - Alireza Farasat
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
2
|
Wei Y, Lv J, Zhu S, Wang S, Su J, Xu C. Enzyme-responsive liposomes for controlled drug release. Drug Discov Today 2024; 29:104014. [PMID: 38705509 DOI: 10.1016/j.drudis.2024.104014] [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: 12/16/2023] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
Compared to other nanovectors, liposomes exhibit unique advantages, such as good biosafety and high drug-loading capacity. However, slow drug release from conventional liposomes makes most payloads unavailable, restricting the therapeutic efficacy. Therefore, in the last ∼20 years, enzyme-responsive liposomes have been extensively investigated, which liberate drugs under the stimulation of enzymes overexpressed at disease sites. In this review, we elaborate on the research progress on enzyme-responsive liposomes. The involved enzymes mainly include phospholipases, particularly phospholipase A2, matrix metalloproteinases, cathepsins, and esterases. These enzymes can cleave ester bonds or specific peptide sequences incorporated in the liposomes for controlled drug release by disrupting the primary structure of liposomes, detaching protective polyethylene glycol shells, or activating liposome-associated prodrugs. Despite decades of efforts, there are still a lack marketed products of enzyme-responsive liposomes. Therefore, more efforts should be made to improve the safety and effectiveness of enzyme-responsive liposomes and address the issues associated with production scale-up.
Collapse
Affiliation(s)
- Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Jiajing Lv
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Shiyu Zhu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Can Xu
- Department of Gastroenterology, Changhai Hospital, Shanghai 200433, China.
| |
Collapse
|
3
|
Fang H, Xu S, Wang Y, Yang H, Su D. Endogenous stimuli-responsive drug delivery nanoplatforms for kidney disease therapy. Colloids Surf B Biointerfaces 2023; 232:113598. [PMID: 37866237 DOI: 10.1016/j.colsurfb.2023.113598] [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: 05/30/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Kidney disease is one of the most life-threatening health problems, affecting millions of people in the world. Commonly used steroids and immunosuppressants often fall exceptionally short of outcomes with inescapable systemic toxicity. With the booming research in nanobiotechnology, stimuli-responsive nanoplatform has come an appealing therapeutic strategy for kidney disease. Endogenous stimuli-responsive materials have shown profuse promise owing to their enhanced spatiotemporal control and precise to the location of the lesion. This review focuses on recent advances stimuli-responsive drug delivery nano-architectonics for kidney disease. First, a brief introduction of pathogenesis of kidney disease and pathological microenvironment were provided. Then, various endogenous stimulus involved in drug delivery nanoplatforms including pH, ROS, enzymes, and glucose were categorized based on the pathological mechanisms of kidney disease. Next, we separately summarized literature examples of endogenous stimuli-responsive nanomaterials, and outlined the design strategies and response mechanisms. Finally, the paper was concluded by discussing remaining challenges and future perspectives of endogenous stimuli-responsive drug delivery nanoplatform for expediting the speed of development and clinical applications.
Collapse
Affiliation(s)
- Hufeng Fang
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China.
| | - Shan Xu
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Yu Wang
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Hao Yang
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Dan Su
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China.
| |
Collapse
|
4
|
Wang Z, Fan X, Mu G, Zhao X, Wang Q, Wang J, Tang X. Cathepsin B-activatable cyclic antisense oligonucleotides for cell-specific target gene knockdown in vitro and in vivo. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:548-558. [PMID: 37588686 PMCID: PMC10425675 DOI: 10.1016/j.omtn.2023.07.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 07/20/2023] [Indexed: 08/18/2023]
Abstract
Trigger-activatable antisense oligonucleotides have been widely applied to regulate gene function. Among them, caged cyclic antisense oligonucleotides (cASOs) maintain a specific topology that temporarily inhibits their interaction with target genes. By inserting linkers that respond to cell-specific endogenous stimuli, they can be powerful tools and potential therapeutic agents for specific types of cancer cells with low off-target effects on normal cells. Here, we developed enzyme-activatable cASOs by tethering two terminals of linear antisense oligonucleotides through a cathepsin B (CB) substrate peptide (Gly-Phe-Leu-Gly [GFLG]), which could be efficiently uncaged by CB. CB-activatable cASOs were used to successfully knock down two disease-related endogenous genes in CB-abundant PC-3 tumor cells at the mRNA and protein levels but had much less effect on gene knockdown in CB-deficient human umbilical vein endothelial cell (HUVECs). In addition, reduced nonspecific immunostimulation was found using cASOs compared with their linear counterparts. Further in vivo studies indicated that CB-activatable cASOs showed effective tumor inhibition in PC-3 tumor model mice through downregulation of translationally controlled tumor protein (TCTP) protein in tumors. This study applies endogenous enzyme-activatable cASOs for antitumor therapy in tumor model mice, which demonstrates a promising stimulus-responsive cASO strategy for cell-specific gene knockdown upon endogenous activation and ASO prodrug development.
Collapse
Affiliation(s)
- Zhongyu Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences and Chemical Biology Center, Peking University, No. 38, Xueyuan Road, Beijing 100191, People’s Republic of China
| | - Xinli Fan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences and Chemical Biology Center, Peking University, No. 38, Xueyuan Road, Beijing 100191, People’s Republic of China
| | - Guanqun Mu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences and Chemical Biology Center, Peking University, No. 38, Xueyuan Road, Beijing 100191, People’s Republic of China
| | - Xiaoran Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences and Chemical Biology Center, Peking University, No. 38, Xueyuan Road, Beijing 100191, People’s Republic of China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences and Chemical Biology Center, Peking University, No. 38, Xueyuan Road, Beijing 100191, People’s Republic of China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences and Chemical Biology Center, Peking University, No. 38, Xueyuan Road, Beijing 100191, People’s Republic of China
| | - Xinjing Tang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences and Chemical Biology Center, Peking University, No. 38, Xueyuan Road, Beijing 100191, People’s Republic of China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu 210023, People’s Republic of China
| |
Collapse
|
5
|
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
|
6
|
Song SJ, Choi JS. Enzyme-Responsive Amphiphilic Peptide Nanoparticles for Biocompatible and Efficient Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14010143. [PMID: 35057039 PMCID: PMC8779831 DOI: 10.3390/pharmaceutics14010143] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 02/03/2023] Open
Abstract
Self-assembled peptide nanostructures recently have gained much attention as drug delivery systems. As biomolecules, peptides have enhanced biocompatibility and biodegradability compared to polymer-based carriers. We introduce a peptide nanoparticle system containing arginine, histidine, and an enzyme-responsive core of repeating GLFG oligopeptides. GLFG oligopeptides exhibit specific sensitivity towards the enzyme cathepsin B that helps effective controlled release of cargo molecules in the cytoplasm. Arginine can induce cell penetration, and histidine facilitates lysosomal escape by its buffering capacity. Herein, we propose an enzyme-responsive amphiphilic peptide delivery system (Arg-His-(Gly-Phe-Lue-Gly)3, RH-(GFLG)3). The self-assembled RH-(GFLG)3 globular nanoparticle structure exhibited a positive charge and formulation stability for 35 days. Nile Red-tagged RH-(GFLG)3 nanoparticles showed good cellular uptake compared to the non-enzyme-responsive control groups with d-form peptides (LD (LRH-D(GFLG)3), DL (DRH-L(GFLG)3), and DD (DRH-D(GFLG)3). The RH-(GFLG)3 nanoparticles showed negligible cytotoxicity in HeLa cells and human RBCs. To determine the drug delivery efficacy, we introduced the anticancer drug doxorubicin (Dox) in the RH-(GFLG)3 nanoparticle system. LL-Dox exhibited formulation stability, maintaining the physical properties of the nanostructure, as well as a robust anticancer effect in HeLa cells compared to DD-Dox. These results indicate that the enzyme-sensitive RH-(GFLG)3 peptide nanoparticles are promising candidates as drug delivery carriers for biomedical applications.
Collapse
Affiliation(s)
- Su Jeong Song
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea;
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Korea
| | - Joon Sig Choi
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Korea
- Correspondence: ; Tel.: +82-(42)-821-5489
| |
Collapse
|
7
|
Application of Non-Viral Vectors in Drug Delivery and Gene Therapy. Polymers (Basel) 2021; 13:polym13193307. [PMID: 34641123 PMCID: PMC8512075 DOI: 10.3390/polym13193307] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/18/2021] [Indexed: 12/13/2022] Open
Abstract
Vectors and carriers play an indispensable role in gene therapy and drug delivery. Non-viral vectors are widely developed and applied in clinical practice due to their low immunogenicity, good biocompatibility, easy synthesis and modification, and low cost of production. This review summarized a variety of non-viral vectors and carriers including polymers, liposomes, gold nanoparticles, mesoporous silica nanoparticles and carbon nanotubes from the aspects of physicochemical characteristics, synthesis methods, functional modifications, and research applications. Notably, non-viral vectors can enhance the absorption of cargos, prolong the circulation time, improve therapeutic effects, and provide targeted delivery. Additional studies focused on recent innovation of novel synthesis techniques for vector materials. We also elaborated on the problems and future research directions in the development of non-viral vectors, which provided a theoretical basis for their broad applications.
Collapse
|
8
|
Ravula V, Lo YL, Wang LF, Patri SV. Gemini Lipopeptide Bearing an Ultrashort Peptide for Enhanced Transfection Efficiency and Cancer-Cell-Specific Cytotoxicity. ACS OMEGA 2021; 6:22955-22968. [PMID: 34514266 PMCID: PMC8427783 DOI: 10.1021/acsomega.1c03620] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/13/2021] [Indexed: 05/08/2023]
Abstract
Cationic gemini lipopeptides are a relatively new class of amphiphilic compounds to be used for gene delivery. Through the possibility of incorporating short peptides with cell-penetrating functionalities, these lipopeptides may be advantageous over traditional cationic lipids. Herein, we report the design, synthesis, and application of a novel cationic gemini lipopeptide for gene delivery. An ultrashort peptide, containing four amino acids, arginine-cysteine-cysteine-arginine, serves as a cationic head group, and two α-tocopherol moieties act as hydrophobic anchoring groups. The new lipopeptide (ATTA) is incorporated into the conventional liposomes, containing 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and 1,2-dioleoyl-sn-glycerol-3-phosphoethanolamine (DOPE), at different molar ratios. The formulated liposomes are characterized and screened for better transfection efficiency. Transfection activity in multiple human cell lines from cancerous and noncancerous origins indicates that the inclusion of an optimal ratio of ATTA in the liposomes substantially enhances the transfection efficiency, superior to that of a traditional liposome, DOTAP-DOPE. Cytotoxicity of ATTA-containing formulations against multiple cell lines indicates potentially distinct activity between cancer and noncancer cell lines. Furthermore, lipoplexes of the ATTA-containing formulations with anticancer therapeutic gene, plasmid encoding tumor necrosis factor-related apoptosis-inducing ligand (pTRAIL), induce obviously more cytotoxicity than conventional formulations. The results indicate that arginine-rich cationic lipopeptide appears to be a promising ingredient in gene delivery vector formulations to enhance transfection efficiency and cell-selective cytotoxicity.
Collapse
Affiliation(s)
- Venkatesh Ravula
- Department
of Chemistry, National Institute of Technology, Warangal 506004, India
- Department
of Medicinal and Applied Chemistry, Kaohsiung
Medical University, Kaohsiung 80708, Taiwan
| | - Yu-Lun Lo
- Department
of Medicinal and Applied Chemistry, Kaohsiung
Medical University, Kaohsiung 80708, Taiwan
| | - Li-Fang Wang
- Department
of Medicinal and Applied Chemistry, Kaohsiung
Medical University, Kaohsiung 80708, Taiwan
- Department
of Medical Research, Kaohsiung Medical University
Hospital, Kaohsiung 80708, Taiwan
| | - Srilakshmi V. Patri
- Department
of Chemistry, National Institute of Technology, Warangal 506004, India
| |
Collapse
|
9
|
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: 36] [Impact Index Per Article: 9.0] [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
|
10
|
Sonju JJ, Dahal A, Singh SS, Jois SD. Peptide-functionalized liposomes as therapeutic and diagnostic tools for cancer treatment. J Control Release 2021; 329:624-644. [PMID: 33010333 PMCID: PMC8082750 DOI: 10.1016/j.jconrel.2020.09.055] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/26/2022]
Abstract
Clinically efficacious medication in anticancer therapy has been successfully designed with liposome-based nanomedicine. The liposomal formulation in cancer drug delivery can be facilitated with a functionalized peptide that mediates the specific drug delivery opportunities with increased drug penetrability, specific accumulation in the targeted site, and enhanced therapeutic efficacy. This review aims to focus on recent advances in peptide-functionalized liposomal formulation techniques in cancer diagnosis and treatment regarding recently published literature. It also will highlight different aspects of novel liposomal formulation techniques that incorporate surface functionalization with peptides for better anticancer effect and current challenges in peptide-functionalized liposomal drug formulation.
Collapse
Affiliation(s)
- Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Seetharama D Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| |
Collapse
|
11
|
Almeida B, Nag OK, Rogers KE, Delehanty JB. Recent Progress in Bioconjugation Strategies for Liposome-Mediated Drug Delivery. Molecules 2020; 25:E5672. [PMID: 33271886 PMCID: PMC7730700 DOI: 10.3390/molecules25235672] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023] Open
Abstract
In nanoparticle (NP)-mediated drug delivery, liposomes are the most widely used drug carrier, and the only NP system currently approved by the FDA for clinical use, owing to their advantageous physicochemical properties and excellent biocompatibility. Recent advances in liposome technology have been focused on bioconjugation strategies to improve drug loading, targeting, and overall efficacy. In this review, we highlight recent literature reports (covering the last five years) focused on bioconjugation strategies for the enhancement of liposome-mediated drug delivery. These advances encompass the improvement of drug loading/incorporation and the specific targeting of liposomes to the site of interest/drug action. We conclude with a section highlighting the role of bioconjugation strategies in liposome systems currently being evaluated for clinical use and a forward-looking discussion of the field of liposomal drug delivery.
Collapse
Affiliation(s)
- Bethany Almeida
- American Society for Engineering Education, Washington, DC 20036, USA;
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
| | - Okhil K. Nag
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
| | - Katherine E. Rogers
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
- Fischell Department of Bioengineering, 2330 Kim Engineering Building, University of Maryland, College Park, MD 20742, USA
| | - James B. Delehanty
- Center for Bio/Molecular Science and Engineering, Code 6900, U.S. Naval Research Laboratory, Washington, DC 20375, USA; (O.K.N.); (K.E.R.)
| |
Collapse
|
12
|
Cathepsin B-Responsive Liposomes for Controlled Anticancer Drug Delivery in Hep G2 Cells. Pharmaceutics 2020; 12:pharmaceutics12090876. [PMID: 32937915 PMCID: PMC7558574 DOI: 10.3390/pharmaceutics12090876] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/17/2022] Open
Abstract
In recent decades, several types of anticancer drugs that inhibit cancer cell growth and cause cell death have been developed for chemotherapeutic application. However, these agents are usually associated with side effects resulting from nonspecific delivery, which may induce cytotoxicity in healthy cells. To reduce the nonspecific delivery issue, nanoparticles have been successfully used for the delivery of anticancer drugs to specific target sites. In this study, a functional polymeric lipid, PEG-GLFG-K(C16)2 (PEG-GLFG, polyethylene glycol-Gly-Leu-Phe-Gly-Lys(C16)2), was synthesized to enable controlled anticancer drug delivery using cathepsin B enzyme-responsive liposomes. The liposomes composed of PEG-GLFG/DOTAP (1,2-dioleoyl-3-trimethylammonium-propane (chloride salt))/DPPC (dipalmitoylphosphatidylcholine)/cholesterol were prepared and characterized at various ratios. The GLFG liposomes formed were stable liposomes and were degraded when acted upon by cathepsin B enzyme. Doxorubicin (Dox) loaded GLFG liposomes (GLFG/Dox) were observed to exert an effective anticancer effect on Hep G2 cells in vitro and inhibit cancer cell proliferation in a zebrafish model.
Collapse
|
13
|
|
14
|
|
15
|
Merino M, Zalba S, Garrido MJ. Immunoliposomes in clinical oncology: State of the art and future perspectives. J Control Release 2018; 275:162-176. [DOI: 10.1016/j.jconrel.2018.02.015] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/09/2018] [Accepted: 02/10/2018] [Indexed: 02/02/2023]
|
16
|
Lee Y, Thompson DH. Stimuli-responsive liposomes for drug delivery. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:10.1002/wnan.1450. [PMID: 28198148 PMCID: PMC5557698 DOI: 10.1002/wnan.1450] [Citation(s) in RCA: 283] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 12/25/2022]
Abstract
The ultimate goal of drug delivery is to increase the bioavailability and reduce the toxic side effects of the active pharmaceutical ingredient (API) by releasing them at a specific site of action. In the case of antitumor therapy, association of the therapeutic agent with a carrier system can minimize damage to healthy, nontarget tissues, while limit systemic release and promoting long circulation to enhance uptake at the cancerous site due to the enhanced permeation and retention effect (EPR). Stimuli-responsive systems have become a promising way to deliver and release payloads in a site-selective manner. Potential carrier systems have been derived from a wide variety of materials, including inorganic nanoparticles, lipids, and polymers that have been imbued with stimuli-sensitive properties to accomplish triggered release based on an environmental cue. The unique features in the tumor microenvironment can serve as an endogenous stimulus (pH, redox potential, or unique enzymatic activity) or the locus of an applied external stimulus (heat or light) to trigger the controlled release of API. In liposomal carrier systems triggered release is generally based on the principle of membrane destabilization from local defects within bilayer membranes to effect release of liposome-entrapped drugs. This review focuses on the literature appearing between November 2008-February 2016 that reports new developments in stimuli-sensitive liposomal drug delivery strategies using pH change, enzyme transformation, redox reactions, and photochemical mechanisms of activation. WIREs Nanomed Nanobiotechnol 2017, 9:e1450. doi: 10.1002/wnan.1450 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Y Lee
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - D H Thompson
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
17
|
Ramasamy T, Ruttala HB, Gupta B, Poudel BK, Choi HG, Yong CS, Kim JO. Smart chemistry-based nanosized drug delivery systems for systemic applications: A comprehensive review. J Control Release 2017; 258:226-253. [PMID: 28472638 DOI: 10.1016/j.jconrel.2017.04.043] [Citation(s) in RCA: 291] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 04/28/2017] [Accepted: 04/30/2017] [Indexed: 12/21/2022]
|
18
|
Zhou Z, Liu X, Zhu D, Wang Y, Zhang Z, Zhou X, Qiu N, Chen X, Shen Y. Nonviral cancer gene therapy: Delivery cascade and vector nanoproperty integration. Adv Drug Deliv Rev 2017; 115:115-154. [PMID: 28778715 DOI: 10.1016/j.addr.2017.07.021] [Citation(s) in RCA: 294] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Gene therapy represents a promising cancer treatment featuring high efficacy and limited side effects, but it is stymied by a lack of safe and efficient gene-delivery vectors. Cationic polymers and lipid-based nonviral gene vectors have many advantages and have been extensively explored for cancer gene delivery, but their low gene-expression efficiencies relative to viral vectors limit their clinical translations. Great efforts have thus been devoted to developing new carrier materials and fabricating functional vectors aimed at improving gene expression, but the overall efficiencies are still more or less at the same level. This review analyzes the cancer gene-delivery cascade and the barriers, the needed nanoproperties and the current strategies for overcoming these barriers, and outlines PEGylation, surface-charge, size, and stability dilemmas in vector nanoproperties to efficiently accomplish the cancer gene-delivery cascade. Stability, surface, and size transitions (3S Transitions) are proposed to resolve those dilemmas and strategies to realize these transitions are comprehensively summarized. The review concludes with a discussion of the future research directions to design high-performance nonviral gene vectors.
Collapse
Affiliation(s)
- Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xiangrui Liu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Dingcheng Zhu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Yue Wang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Zhen Zhang
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuefei Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Nasha Qiu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China
| | - Xuesi Chen
- Changchun Institute of Applied Chemistry, Key Lab of Polymer Ecomaterials, Changchun, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027 Hangzhou, China.
| |
Collapse
|
19
|
Mathew AP, Cho KH, Uthaman S, Cho CS, Park IK. Stimuli-Regulated Smart Polymeric Systems for Gene Therapy. Polymers (Basel) 2017; 9:E152. [PMID: 30970831 PMCID: PMC6432211 DOI: 10.3390/polym9040152] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 01/02/2023] Open
Abstract
The physiological condition of the human body is a composite of different environments, each with its own parameters that may differ under normal, as well as diseased conditions. These environmental conditions include factors, such as pH, temperature and enzymes that are specific to a type of cell, tissue or organ or a pathological state, such as inflammation, cancer or infection. These conditions can act as specific triggers or stimuli for the efficient release of therapeutics at their destination by overcoming many physiological and biological barriers. The efficacy of conventional treatment modalities can be enhanced, side effects decreased and patient compliance improved by using stimuli-responsive material that respond to these triggers at the target site. These stimuli or triggers can be physical, chemical or biological and can be internal or external in nature. Many smart/intelligent stimuli-responsive therapeutic gene carriers have been developed that can respond to either internal stimuli, which may be normally present, overexpressed or present in decreased levels, owing to a disease, or to stimuli that are applied externally, such as magnetic fields. This review focuses on the effects of various internal stimuli, such as temperature, pH, redox potential, enzymes, osmotic activity and other biomolecules that are present in the body, on modulating gene expression by using stimuli-regulated smart polymeric carriers.
Collapse
Affiliation(s)
- Ansuja Pulickal Mathew
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Ki-Hyun Cho
- Department of Plastic Surgery, Institute of Dermatology and Plastic Surgery, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA.
| | - Saji Uthaman
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.
| | - In-Kyu Park
- Department of Biomedical Sciences, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Korea.
| |
Collapse
|