1
|
Ho KW, Liu YL, Huang BC, Hong ST, Yang SH, Liao TY, Liu ES, Chen YT, Huang YZ, Leu YL, Chen CY, Chen BM, Roffler SR, Cheng TL. Targeted internalization and activation of glycosidic switch liposomes by a biological macromolecule mPEG×EphA2 increases therapeutic efficacy against lung cancer. Int J Biol Macromol 2025; 300:140138. [PMID: 39842595 DOI: 10.1016/j.ijbiomac.2025.140138] [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: 09/05/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Glycosidic switch liposome (GSL) technology efficiently encapsulates and stabilizes potent anticancer drugs in liposomes using a reversible glucuronide ester. Enzymatic hydrolysis of the glucuronide switch in target cell lysosomes produces parental drug. Our study examined the potential of a bispecific macromolecule, a polyethylene glycol (PEG) engager (mPEG×EphA2), generated by fusing a humanized anti-methoxy PEG (mPEG) Fab with an anti-EphA2 single-chain antibody, to increase GSL uptake into cancer cells and boost the anticancer activity by targeting PEG on GSL and an internalizing tumor antigen. Combining GSL with the PEG engager creates αEphA2/GSL, targeting cancer cells to generate topoisomerase I poison 9-aminocamptothecin (9 AC) for cell killing. Targeted liposomes can bind CL1-5 human lung adenocarcinoma cells and increase GSL internalization from 0 % to 62.4 % in 60 min. αEphA2/GSL showed slightly higher cellular cytotoxicity than non-targeted GSL, but targeted GSL increased 9 AC intratumoral concentrations by 8.4 fold at 24 h. The 9 AC tumor/blood ratio of αEphA2/GSL was nearly 6-fold higher than αDNS/GSL (control engager GSL). Using targeted GSL, five of seven mice with solid CL1-5 tumors were cured. The mPEG×EphA2 engager can enhance GSL drug uptake and generation, boosting lung cancer treatment efficacy, suggesting that αEphA2/GSL is a promising treatment for tumors overexpressing EphA2.
Collapse
Affiliation(s)
- Kai-Wen Ho
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Ling Liu
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bo-Cheng Huang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Shih-Ting Hong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Hung Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taiwan
| | - Tzu-Yi Liao
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - En-Shuo Liu
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Tung Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Zhong Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Lin Leu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Chiao-Yun Chen
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steve R Roffler
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
2
|
Torres J, Valenzuela Oses JK, Rabasco-Álvarez AM, González-Rodríguez ML, García MC. Innovations in Cancer Therapy: Endogenous Stimuli-Responsive Liposomes as Advanced Nanocarriers. Pharmaceutics 2025; 17:245. [PMID: 40006612 PMCID: PMC11858840 DOI: 10.3390/pharmaceutics17020245] [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: 12/21/2024] [Revised: 02/03/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Recent advancements in nanotechnology have revolutionized cancer therapy-one of the most pressing global health challenges and a leading cause of death-through the development of liposomes (L), lipid-based nanovesicles known for their biocompatibility and ability to encapsulate both hydrophilic and lipophilic drugs. More recent innovations have led to the creation of stimuli-responsive L that release their payloads in response to specific endogenous or exogenous triggers. Dual- and multi-responsive L, which react to multiple stimuli, offer even greater precision, improving therapeutic outcomes while reducing systemic toxicity. Additionally, these smart L can adjust their physicochemical properties and morphology to enable site-specific targeting and controlled drug release, enhancing treatment efficacy while minimizing adverse effects. This review explores the latest advancements in endogenous stimuli-responsive liposomal nanocarriers, as well as dual- and multi-responsive L that integrate internal and external triggers, with a focus on their design strategies, mechanisms, and applications in cancer therapy.
Collapse
Affiliation(s)
- Jazmín Torres
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, Science Building 2, Córdoba X5000HUA, Argentina; (J.T.); (J.K.V.O.)
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Córdoba X5000HUA, Argentina
| | - Johanna Karina Valenzuela Oses
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, Science Building 2, Córdoba X5000HUA, Argentina; (J.T.); (J.K.V.O.)
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Córdoba X5000HUA, Argentina
| | - Antonio María Rabasco-Álvarez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain;
| | - María Luisa González-Rodríguez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain;
| | - Mónica Cristina García
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, Science Building 2, Córdoba X5000HUA, Argentina; (J.T.); (J.K.V.O.)
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Córdoba X5000HUA, Argentina
| |
Collapse
|
3
|
Elsayed N. Selective imaging, gene, and therapeutic delivery using PEGylated and pH-Sensitive nanoparticles for enhanced lung disorder treatment. Int J Pharm 2024; 666:124819. [PMID: 39424084 DOI: 10.1016/j.ijpharm.2024.124819] [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: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Lung inflammation involves the activation of immune cells and inflammatory mediators in response to injury and infection. When inflammation persists, fibroblasts, which are resident lung cells, become activated, leading to pulmonary fibrosis (PF), abnormal wound healing, and long-term damage to the alveolar epithelium. This persistent inflammation and fibrosis can also elevate the risk of lung cancer, emphasizing the need for innovative treatments. Current therapies, such as inhaled corticosteroids (ICS) and chemotherapy, have significant limitations. Although conventional nanoparticles (NPs) provide a promising avenue for treating lung disorders, they have limited selectivity and stability. Polyethylene glycol (PEG) grafting can prevent NP aggregation and phagocytosis, thus prolonging their circulation time. When combined with targeting ligands, PEGylated NPs can deliver drugs precisely to specific cells or tissues. Moreover, pH-sensitive NPs offer the advantage of selective drug delivery to inflammatory or tumor-acidic environments, reducing side effects. These NPs can change their size, shape, or surface charge in response to pH variations, improving drug delivery efficiency. This review examines the techniques of PEGylation, the polymers used in pH-sensitive NPs, and their therapeutic applications for lung inflammation, fibrosis, and cancer. By harnessing innovative NP technologies, researchers can develop effective therapies for respiratory conditions, addressing unmet medical needs and enhancing patient outcomes.
Collapse
Affiliation(s)
- Nourhan Elsayed
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, Subang Jaya, Malaysia.
| |
Collapse
|
4
|
Mirhadi E, Askarizadeh A, Farhoudi L, Mashreghi M, Behboodifar S, Alavizadeh SH, Arabi L, Jaafari MR. The impact of phospholipids with high transition temperature to enhance redox-sensitive liposomal doxorubicin efficacy in colon carcinoma model. Chem Phys Lipids 2024; 261:105396. [PMID: 38621603 DOI: 10.1016/j.chemphyslip.2024.105396] [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: 02/12/2024] [Revised: 04/01/2024] [Accepted: 04/11/2024] [Indexed: 04/17/2024]
Abstract
In this study, we have developed a redox-sensitive (RS) liposomal doxorubicin formulation by incorporating 10,10'-diselanediylbis decanoic acid (DDA) organoselenium compound as the RS moiety. Hence, several RS liposomal formulations were prepared by using DOPE, HSPC, DDA, mPEG2000-DSPE, and cholesterol. In situ drug loading using a pH gradient and citrate complex yielded high drug to lipid ratio and encapsulation efficiency (100%) for RS liposomes. Liposomal formulations were characterized in terms of size, surface charge and morphology, drug loading, release properties, cell uptake and cytotoxicity, as well as therapeutic efficacy in BALB/c mice bearing C26 tumor cells. The formulations showed an average particle size of 200 nm with narrow size distributions (PDI < 0.3), and negative surface charges varying from -6 mV to -18.6 mV. Our study confirms that the presence of the DDA compound in liposomes is highly sensitive to hydrogen peroxide at 0.1% w/v, resulting in a significant burst release of up to 40%. The in vivo therapeutic efficacy study in BALB/c mice bearing C26 colon carcinoma confirmed the promising function of RS liposomes in the tumor microenvironment which led to a prolonged median survival time (MST). The addition of hydrogenated soy phosphatidylcholine (HSPC) with a high transition temperature (Tm: 52-53.5°C) extended the MST of our 3-component formulation of F14 (DOPE/HSPC/DDA) to 60 days in comparison to Caelyx (PEGylated liposomal Dox), which is not RS-sensitive (39 days). Overall, HSPC liposomes bearing RS-sensitive moiety enhanced therapeutic efficacy against colon cancer in vitro and in vivo. This achievement unequivocally underscores the criticality of high-TM phospholipids, particularly HSPC, in significantly enhancing liposome stability within the bloodstream. In addition, RS liposomes enable the on-demand release of drugs, leveraging the redox environment of tumor cells, thereby augmenting the efficacy of the formulation.
Collapse
Affiliation(s)
- Elaheh Mirhadi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Anis Askarizadeh
- Marine Pharmaceutical Science Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Leila Farhoudi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Mashreghi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Behboodifar
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Nasri N, Saharkhiz S, Dini G, Yousefnia S. Thermo- and pH-responsive targeted lipid-coated mesoporous nano silica platform for dual delivery of paclitaxel and gemcitabine to overcome HER2-positive breast cancer. Int J Pharm 2023; 648:123606. [PMID: 37972671 DOI: 10.1016/j.ijpharm.2023.123606] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/09/2023] [Accepted: 11/11/2023] [Indexed: 11/19/2023]
Abstract
In the current study, a new monoclonal antibody conjugated dual stimuli lipid-coated mesoporous silica nanoparticles (L-MSNs) platform was developed and investigated for specific co-delivery of the paclitaxel (PTX) and gemcitabine (Gem) to cancer cells and preventing their side effects during the treatment process. First, MSNs were synthesized and then coated with as-prepared pH-, and thermo-sensitive niosomes to produce L-MSNs. For this aim, Dipalmitoylphosphatidylcholine (DPPC) was used to create thermo-sensitivity, and 1, 2-Distearoyl-sn-glycerol-3-phosphoethanolamine -Citraconic Anhydride-Polyethylene Glycol (DSPE-CA-PEG) polymers were prepared and incorporated to the lipid layer for creation of pH-sensitivity. In the next step, trastuzumab as a monoclonal antibody (mAb) was conjugated to the maleimide groups of the 1, 2-Distearoyl-sn-glycerol-3-phosphoethanolamine DSPE-polyethylene glycol (PEG)-maleimide agents in the lipid bilayer via a disulfide bond. Dynamic light scattering (DLS) and zeta potential measurements, Fourier-transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), Brunauer-Emmett-Teller (BET), and scanning electron microscopy (SEM) analyses were utilized to characterize the synthesized particles before and after surface modification. The encapsulation efficiency (EE%) and loading efficiency (LE%) of the particles were also evaluated. Additionally, the drug release study and MTT assay were done to evaluate the bioactivity potential of the fabricated platforms. The results of DLS and zeta potential measurements revealed an average size of 200 nm and a neutral zeta potential of about -1 mV for mAb-L-MSNs. Also, the FTIR spectra confirmed the formation of mAb-L-MSNs. Moreover, SEM analysis showed spherical-shaped MSNs with amorphous structure confirmed by XRD analysis, and BET test revealed ∼ 820 m2/g specific surface area and pore about 5 nm in size. The values of EE% and LE% of PTX were 90.3 % and 26.7 %, while these values for GEM were 89.5 % and 38.8 % in the co-loaded form, respectively. The thermo-pH-sensitivity examination showed approximately 500 nm of size increase after the change of pH and temperature from 7.4 and 37˚C to 5 and 42˚C. The release profile showed a pH-, and thermo-dependence manner, which led to about 89 % and 95 % of PTX and GEM released from the co-loaded platform at a pH of 5 and 42 °C while these values were 31.1 % and 32.2 % at pH of 7.4 and 37˚C, respectively. MTT assay data presented that when the mAb-L-co-loaded-MSNs platform containing 250 µg/mL drug was used, about 92 % of cells died in human epidermal receptors (HER2)-positive breast cancer cells (SKBR3), while just about 4 % of HER2-negative normal cells were killed. However, the growth inhibition rate of SKBR3 cells was caused by empty-mAb-L-MSNs, pure PTX and GEM combination were 9 % and 87 %, respectively. Moreover, the half inhibitory concentration (IC50) of the pure PTX, pure GEM, and mAb-coloaded-L-MSNs were 33, 17.6, and 6.5 µg/mL. The synergic effect of co-encapsulation of PTX and GEM in addition to trastuzumab conjugated L-MSNs was confirmed by a combinational index (CI) of 0.34. Therefore, this strategy leads to specific targeted drug delivery to cancer cells using a key-lock interaction between the trastuzumab and HER-2 receptors on the cancer cell membrane which stimuli the endocytosis of the particles to the cells followed by the destruction of the lipid layer in the acidic pH and the temperature of the lysosome, leading to enhanced release of PTX and GEM (pH of 5 and 42˚C). So, this platform can be considered a suitable carrier for cancer treatment.
Collapse
Affiliation(s)
- Negar Nasri
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Shaghayegh Saharkhiz
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Ghasem Dini
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran.
| | - Saghar Yousefnia
- Department of Cell and Molecular Biology, Semnan University, Semnan, Iran
| |
Collapse
|
6
|
Liu C, Wu K, Li J, Mu X, Gao H, Xu X. Nanoparticle-mediated therapeutic management in cholangiocarcinoma drug targeting: Current progress and future prospects. Biomed Pharmacother 2023; 158:114135. [PMID: 36535198 DOI: 10.1016/j.biopha.2022.114135] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Patients with cholangiocarcinoma (CCA) often have an unfavorable prognosis because of its insidious nature, low resectability rate, and poor response to anticancer drugs and radiotherapy, which makes early detection and treatment difficult. At present, CCA has a five-year overall survival rate (OS) of only 5%, despite advances in therapies. New an increasing number of evidence suggests that nanoplatforms may play a crucial role in enhancing the pharmacological effects and in reducing both short- and long-term side effects of cancer treatment. This document reviews the advantages and shortcomings of nanoparticles such as liposomes, polymeric nanoparticle,inorganic nanoparticle, nano-metals and nano-alloys, carbon dots, nano-micelles, dendrimer, nano-capsule, bio-Nanomaterials in the diagnosis and treatment of CCA and discuss the current challenges in of nanoplatforms for CCA.
Collapse
Affiliation(s)
- Chunkang Liu
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Kunzhe Wu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jianyang Li
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xupeng Mu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Huan Gao
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaohua Xu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
7
|
Ge X, Hu Y, Shen H, Liang W, Sun Z, Zhang X, Li W. Pheophorbide-a as a Light-Triggered Liposomal Switch: For the Controlled Release of Alpinia galanga ( A. galanga) Essential Oil and Its Stability, Antioxidant, and Antibacterial Activity Assessment. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1667-1678. [PMID: 36629793 DOI: 10.1021/acs.jafc.2c07082] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In this study, Alpinia galanga essential oil liposomes (EO-Lip) were prepared with soybean lecithin and cholesterol as wall materials. A light-responsive liposome (EO-PLip) was designed for the controlled release of A. galanga oil based on the light-responsive properties of Pheophorbide-a. The dependence of Pheophorbide-a on illumination time was proved by UV spectroscopy. Characterization techniques such as UV spectroscopy, transmission electron microscopy, and Fourier transform infrared spectroscopy demonstrated that the essential oils were successfully encapsulated in liposomes. Moreover, the particle size of EO-PLip was 166.30 nm, the polydispersity index was 0.22, the zeta potential was -49.50 mV, and the encapsulation efficiency was 30.83%. Both EO-Lip and EO-Plip have high sustained-release effects on essential oil and showed light-responsive release characteristics under infrared stimulation. The prepared liposomes had good storage stability at 4 °C for 28 d. EO-PLip showed excellent transient antioxidant and bacteriostatic properties based on the ability to respond to light and slow release. This EO-PLip provided a platform for essential oils and might be used as a potent and controllable solution.
Collapse
Affiliation(s)
- Xiangzhen Ge
- Engineering Research Center of Grain and Oil Functionalized Processing in Universities of Shaanxi Province, College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi712100, P. R. China
- Key Laboratory of Agro-products Quality and Safety Controlling Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Beijing100193, P. R. China
| | - Yayun Hu
- Engineering Research Center of Grain and Oil Functionalized Processing in Universities of Shaanxi Province, College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi712100, P. R. China
| | - Huishan Shen
- Engineering Research Center of Grain and Oil Functionalized Processing in Universities of Shaanxi Province, College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi712100, P. R. China
| | - Wei Liang
- Engineering Research Center of Grain and Oil Functionalized Processing in Universities of Shaanxi Province, College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi712100, P. R. China
| | - Zhuangzhuang Sun
- Engineering Research Center of Grain and Oil Functionalized Processing in Universities of Shaanxi Province, College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi712100, P. R. China
| | - Xiuyun Zhang
- Engineering Research Center of Grain and Oil Functionalized Processing in Universities of Shaanxi Province, College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi712100, P. R. China
| | - Wenhao Li
- Engineering Research Center of Grain and Oil Functionalized Processing in Universities of Shaanxi Province, College of Food Science and Engineering, Northwest A&F University, 22 Xinong Road, Yangling, Shaanxi712100, P. R. China
- Key Laboratory of Agro-products Quality and Safety Controlling Storage and Transport Process, Ministry of Agriculture and Rural Affairs, Beijing100193, P. R. China
| |
Collapse
|
8
|
Updates on Responsive Drug Delivery Based on Liposome Vehicles for Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14102195. [PMID: 36297630 PMCID: PMC9608678 DOI: 10.3390/pharmaceutics14102195] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/20/2022] Open
Abstract
Liposomes are well-known nanoparticles with a non-toxic nature and the ability to incorporate both hydrophilic and hydrophobic drugs simultaneously. As modern drug delivery formulations are produced by emerging technologies, numerous advantages of liposomal drug delivery systems over conventional liposomes or free drug treatment of cancer have been reported. Recently, liposome nanocarriers have exhibited high drug loading capacity, drug protection, improved bioavailability, enhanced intercellular delivery, and better therapeutic effect because of resounding success in targeting delivery. The site targeting of smart responsive liposomes, achieved through changes in their physicochemical and morphological properties, allows for the controlled release of active compounds under certain endogenous or exogenous stimuli. In that way, the multifunctional and stimuli-responsive nanocarriers for the drug delivery of cancer therapeutics enhance the efficacy of treatment prevention and fighting over metastases, while limiting the systemic side effects on healthy tissues and organs. Since liposomes constitute promising nanocarriers for site-targeted and controlled anticancer drug release, this review focuses on the recent progress of smart liposome achievements for anticancer drug delivery applications.
Collapse
|
9
|
Li J, Gao Y, Liu S, Cai J, Zhang Q, Li K, Liu Z, Shi M, Wang J, Cui H. Aptamer-functionalized Quercetin Thermosensitive Liposomes for Targeting Drug Delivery and Antitumor Therapy. Biomed Mater 2022; 17. [PMID: 36001994 DOI: 10.1088/1748-605x/ac8c75] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/24/2022] [Indexed: 11/12/2022]
Abstract
Chemo-thermotherapy, as a promising cancer combination therapy strategy, has attracted widespread attention. In this study, a novel aptamer functionalized thermosensitive liposome encapsulating hydrophobic drug quercetin was fabricated as an efficient drug delivery system. This aptamer-functionalized quercetin thermosensitive liposomes (AQTSL) combined the merits of high-loading yield, sustained drug release, long-term circulation in the body of PEGylated liposomes, passive targeting provided by 100-200 nm nanoparticles, active targeting and improved internalization effects offered by AS1411 aptamer, and temperature-responsive of quercetin release. In addition, AQTSL tail vein injection combined with 42℃ water bath heating on tumor site (AQTSL+42℃)treatment inhibited the tumor growth significantly compared with the normal saline administration (p<0.01), and the inhibition rate reached 75%. Furthermore, AQTSL+42℃ treatment also slowed down the tumor growth significantly compared with QTSL combined with 42℃ administration (p<0.05), confirming that AS1411 decoration on QTSL increased the active targeting and internalization effects of the drug delivery system, and AS1411 aptamer itself might also contribute to the tumor inhibition. These data indicate that AQTSL is a potential carrier candidate for different hydrophobic drugs and tumor targeting delivery, and this kind of targeted drug delivery system combined with temperature responsive drug release mode is expected to achieve an ideal tumor therapy effect.
Collapse
Affiliation(s)
- Jian Li
- Yanshan University, No.438,Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Yanting Gao
- Yanshan University, No.438, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Shihe Liu
- Yanshan University, No.438,Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Jiahui Cai
- Yanshan University, No.438, Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Qing Zhang
- Yanshan University, No.438, Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Kun Li
- Yanshan University, No. 438, Hebei Street, Qinhuangdao, Hebei Province, 066000, CHINA
| | - Zhiwei Liu
- Yanshan University, No. 438, West Section of Hebei Street, Qinhuangdao, Hebei, 066004, CHINA
| | - Ming Shi
- Yanshan University, No.438, Hebei Street, Qinhuangdao, Hebei Province, 066004, CHINA
| | - Jidong Wang
- Yanshan University, No. 438, Hebei Street, Qinhuangdao, 066000, CHINA
| | - Hongxia Cui
- Yanshan University, No. 438, Hebei Street, Qinhuangdao, Hebei Province, 066004, CHINA
| |
Collapse
|
10
|
Lamparelli EP, Ciardulli MC, Scala P, Scognamiglio M, Charlier B, Di Pietro P, Izzo V, Vecchione C, Maffulli N, Della Porta G. Lipid nano-vesicles for thyroid hormone encapsulation: A comparison between different fabrication technologies, drug loading, and an in vitro delivery to human tendon stem/progenitor cells in 2D and 3D culture. Int J Pharm 2022; 624:122007. [PMID: 35820518 DOI: 10.1016/j.ijpharm.2022.122007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 06/06/2022] [Accepted: 07/05/2022] [Indexed: 02/08/2023]
Abstract
Phosphatidylcholine (PC) vesicles loaded with Triiodothyronine (T3) were fabricated using different manufacturing methods: thin layer hydration plus sonication (TF-UF), supercritical liposome formation (SC), and microfluidic technology (MF). Vesicles obtained by MF had the lowest mean diameter (88.61 ± 44.48 nm) with a Zeta Potential of -20.1 ± 5.90 mV and loading of 10 mg/g (encapsulation efficiency: 57%). In contrast, SC vesicles showed extremely low encapsulation efficiency (<10%) probably due to T3 solubility in ethanol/carbon dioxide mixture; despite TF-UF vesicles exhibiting good size (167.7 ± 90 nm; Zp -8.50 ± 0.60 mV) and loading (10 mg/g), poor mass recovery was obtained (50% loss). MF vesicles had low cytotoxicity, and they were well enough internalized by both HeLa and human tendon stem/progenitor cells (hTSPCs). Their biological activity was also monitored in both 2D and 3D cultures of hTSPCs supplemented with therapeutical concentrations of PC/T3 nano-liposomes. 2D culture showed almost similar constitutive gene expression compared to control culture supplemented with free-T3. On the contrary, when hTPSCs 3D culture was assembled, it showed a more evident homogeneous distribution of FITC labeled vesicles within the high-density structure and a significant upregulation of cell constitutive genes, such as type I Collagen (4.8-fold; p < 0.0001) at day 7, compared to the control, suggesting that T3/PC formulation has increased T3 cytosolic concentration, thus improving cells metabolic activity. The study supported MF technology for nano-carriers fabrication and opens perspectives on the activity of PC/T3 nano-vesicles as innovative formulations for TPSCs stimulation in ECM secretion.
Collapse
Affiliation(s)
- E P Lamparelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, (SA), Italy
| | - M C Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, (SA), Italy
| | - P Scala
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, (SA), Italy
| | - M Scognamiglio
- Department of Industrial Engineering, Università di Salerno, via Giovanni Paolo I, 84084 Fisciano, (SA), Italy
| | - B Charlier
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, (SA), Italy
| | - P Di Pietro
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, (SA), Italy
| | - V Izzo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, (SA), Italy
| | - C Vecchione
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, (SA), Italy; IRCCS Neuromed, Department of Vascular Physiopathology, 86077 Pozzilli, IS, Italy
| | - N Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, (SA), Italy
| | - G Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, (SA), Italy; Department of Industrial Engineering, Università di Salerno, via Giovanni Paolo I, 84084 Fisciano, (SA), Italy; Interdepartment Centre BIONAM, Università di Salerno, via Giovanni Paolo I, 84084 Fisciano, (SA), Italy.
| |
Collapse
|
11
|
Preparation and application of pH-responsive drug delivery systems. J Control Release 2022; 348:206-238. [PMID: 35660634 DOI: 10.1016/j.jconrel.2022.05.056] [Citation(s) in RCA: 187] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 02/08/2023]
Abstract
Microenvironment-responsive drug delivery systems (DDSs) can achieve targeted drug delivery, reduce drug side effects and improve drug efficacies. Among them, pH-responsive DDSs have gained popularity since the pH in the diseased tissues such as cancer, bacterial infection and inflammation differs from a physiological pH of 7.4 and this difference could be harnessed for DDSs to release encapsulated drugs specifically to these diseased tissues. A variety of synthetic approaches have been developed to prepare pH-sensitive DDSs, including introduction of a variety of pH-sensitive chemical bonds or protonated/deprotonated chemical groups. A myriad of nano DDSs have been explored to be pH-responsive, including liposomes, micelles, hydrogels, dendritic macromolecules and organic-inorganic hybrid nanoparticles, and micron level microspheres. The prodrugs from drug-loaded pH-sensitive nano DDSs have been applied in research on anticancer therapy and diagnosis of cancer, inflammation, antibacterial infection, and neurological diseases. We have systematically summarized synthesis strategies of pH-stimulating DDSs, illustrated commonly used and recently developed nanocarriers for these DDSs and covered their potential in different biomedical applications, which may spark new ideas for the development and application of pH-sensitive nano DDSs.
Collapse
|
12
|
García MC, Calderón-Montaño JM, Rueda M, Longhi M, Rabasco AM, López-Lázaro M, Prieto-Dapena F, González-Rodríguez ML. pH-temperature dual-sensitive nucleolipid-containing stealth liposomes anchored with PEGylated AuNPs for triggering delivery of doxorubicin. Int J Pharm 2022; 619:121691. [PMID: 35331830 DOI: 10.1016/j.ijpharm.2022.121691] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/05/2022] [Accepted: 03/18/2022] [Indexed: 11/29/2022]
Abstract
Liposomes (Lip) are useful nanocarriers for drug delivery and cancer nanomedicine because of their ability to efficiently encapsulate drugs with different physical and chemical properties. The pH gradient between normal and tumoral tissues, and their rapid metabolism that induces hyperthermia encourage the development of pH- and thermo-sensitive Lip for delivering anticancer drugs. Nucleolipids have been studied as scaffolding material to prepare Lip, mainly for cancer therapy. Herein, we report for the first time the use of 1,2-dipalmitoyl-sn-glycero-3-(cytidine diphosphate) (DG-CDP) to develop pH/thermo-sensitive nucleolipid-containing stealth Lip stabilized by combination with 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) and cholesterol, anchored with NH2-PEGylated gold nanoparticles (PEG-AuNPs, 15 nm) for triggering delivery of doxorubicin (Dox). The optimal composition of DPPC, DG-CDP and cholesterol (94:3:3) was established by Langmuir isotherms. Unloaded and Dox-loaded Lip and AuNPs-Lip exhibited nano-scale sizes (415-650 nm), acceptable polydispersity indexes (<0.33), spherical shapes, and negative Z-potential (-23- -6.6 mV) due to the phosphate groups of DG-CDP, which allowed the anchoring with positively charged AuNPs. High EE% were achieved (>78%) and although efficient control in the Dox release towards different receptor media was observed, the release of Dox from PEG-AuNPs-Lip-Dox was significantly triggered at acidic pH and hyperthermia temperature, demonstrating its responsiveness to both stimuli. Dox-loaded Lip showed high cytotoxic activity against MDA-MB-231 breast cancer cells and SK-OV-3 ovarian cancer cells, suggesting that Dox was released from these nanocarriers over time. Overall, the liposomal formulations showed promising properties as stimuli-responsive nanocarriers for cancer nanomedicine, with prospects for hyperthermia therapy.
Collapse
Affiliation(s)
- Mónica C García
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, Ciudad Universitaria, Haya de la Torre and Medina Allende, Science Building 2, Córdoba X5000HUA, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Córdoba X5000HUA, Argentina; Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain.
| | - José Manuel Calderón-Montaño
- Department of Pharmacology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain
| | - Manuela Rueda
- Department of Physical Chemistry, Faculty of Chemistry, Universidad de Sevilla, C/Prof. García González s/n, 41012 Seville, Spain
| | - Marcela Longhi
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Ciencias Farmacéuticas, Ciudad Universitaria, Haya de la Torre and Medina Allende, Science Building 2, Córdoba X5000HUA, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Córdoba X5000HUA, Argentina
| | - Antonio M Rabasco
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain
| | - Miguel López-Lázaro
- Department of Pharmacology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain
| | - Francisco Prieto-Dapena
- Department of Physical Chemistry, Faculty of Chemistry, Universidad de Sevilla, C/Prof. García González s/n, 41012 Seville, Spain
| | - María Luisa González-Rodríguez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain.
| |
Collapse
|
13
|
Gorjian H, Raftani Amiri Z, Mohammadzadeh Milani J, Ghaffari Khaligh N. Influence of Nanovesicle Type, Nanoliposome and Nanoniosome, on Antioxidant and Antimicrobial Activities of Encapsulated Myrtle Extract: A Comparative Study. FOOD BIOPROCESS TECH 2022. [DOI: 10.1007/s11947-021-02747-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
14
|
Amigh S, Mohajeri A. Coronene-based quantum dots for the delivery of the doxorubicin anticancer drug: a computational study. NEW J CHEM 2022. [DOI: 10.1039/d2nj00636g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The coronene family could serve as a useful platform for the delivery of and tracking the release of the anticancer DOX drug.
Collapse
Affiliation(s)
- Soode Amigh
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 7194684795, Iran
| | - Afshan Mohajeri
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz 7194684795, Iran
| |
Collapse
|
15
|
Rahimi R, Solimannejad M, Soleimannejad M. Two-dimensionalcovalent triazine frameworks as superior nanocarriers for the delivery of thioguanine anti-cancer drugs: a periodic DFT study. NEW J CHEM 2022. [DOI: 10.1039/d2nj02050e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This work aims to introduce a superior nanocarrier for thioguanine (TG) anti-cancer drug delivery, drug release, and cancer therapy through computational chemistry.
Collapse
Affiliation(s)
- Rezvan Rahimi
- Department of Chemistry, Faculty of Science, Arak University, Arak 38156-8-8349, Iran
- Institute of Nanosciences and Nanotechnology, Arak University, Arak 38156-8-8349, Iran
| | - Mohammad Solimannejad
- Department of Chemistry, Faculty of Science, Arak University, Arak 38156-8-8349, Iran
- Institute of Nanosciences and Nanotechnology, Arak University, Arak 38156-8-8349, Iran
| | | |
Collapse
|
16
|
Stimuli-responsive nanoliposomes as prospective nanocarriers for targeted drug delivery. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102916] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
17
|
Xu M, Yim W, Zhou J, Zhou J, Jin Z, Moore C, Borum R, Jorns A, Jokerst JV. The Application of Organic Nanomaterials for Bioimaging, Drug Delivery, and Therapy: Spanning Various Domains. IEEE NANOTECHNOLOGY MAGAZINE 2021. [DOI: 10.1109/mnano.2021.3081758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
18
|
AlSawaftah N, Pitt WG, Husseini GA. Dual-Targeting and Stimuli-Triggered Liposomal Drug Delivery in Cancer Treatment. ACS Pharmacol Transl Sci 2021; 4:1028-1049. [PMID: 34151199 PMCID: PMC8205246 DOI: 10.1021/acsptsci.1c00066] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 12/31/2022]
Abstract
The delivery of chemotherapeutics to solid tumors using smart drug delivery systems (SDDSs) takes advantage of the unique physiology of tumors (i.e., disordered structure, leaky vasculature, abnormal extracellular matrix (ECM), and limited lymphatic drainage) to deliver anticancer drugs with reduced systemic side effects. Liposomes are the most promising of such SDDSs and have been well investigated for cancer therapy. To improve the specificity, bioavailability, and anticancer efficacy of liposomes at the diseased sites, other strategies such as targeting ligands and stimulus-sensitive liposomes have been developed. This review highlights relevant surface functionalization techniques and stimuli-mediated drug release for enhanced delivery of anticancer agents at tumor sites, with a special focus on dual functionalization and design of multistimuli responsive liposomes.
Collapse
Affiliation(s)
- Nour AlSawaftah
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, UAE
| | - William G. Pitt
- Chemical
Engineering Department, Brigham Young University, Provo, Utah 84602, United States
| | - Ghaleb A. Husseini
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, UAE
| |
Collapse
|
19
|
De Leo V, Milano F, Agostiano A, Catucci L. Recent Advancements in Polymer/Liposome Assembly for Drug Delivery: From Surface Modifications to Hybrid Vesicles. Polymers (Basel) 2021; 13:1027. [PMID: 33810273 PMCID: PMC8037206 DOI: 10.3390/polym13071027] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Liposomes are consolidated and attractive biomimetic nanocarriers widely used in the field of drug delivery. The structural versatility of liposomes has been exploited for the development of various carriers for the topical or systemic delivery of drugs and bioactive molecules, with the possibility of increasing their bioavailability and stability, and modulating and directing their release, while limiting the side effects at the same time. Nevertheless, first-generation vesicles suffer from some limitations including physical instability, short in vivo circulation lifetime, reduced payload, uncontrolled release properties, and low targeting abilities. Therefore, liposome preparation technology soon took advantage of the possibility of improving vesicle performance using both natural and synthetic polymers. Polymers can easily be synthesized in a controlled manner over a wide range of molecular weights and in a low dispersity range. Their properties are widely tunable and therefore allow the low chemical versatility typical of lipids to be overcome. Moreover, depending on their structure, polymers can be used to create a simple covering on the liposome surface or to intercalate in the phospholipid bilayer to give rise to real hybrid structures. This review illustrates the main strategies implemented in the field of polymer/liposome assembly for drug delivery, with a look at the most recent publications without neglecting basic concepts for a simple and complete understanding by the reader.
Collapse
Affiliation(s)
- Vincenzo De Leo
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy;
| | - Francesco Milano
- Istituto di Scienze delle Produzioni Alimentari (ISPA), Consiglio Nazionale delle Ricerche (CNR), S.P. Lecce-Monteroni, Ecotekne, 73100 Lecce, Italy;
| | - Angela Agostiano
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy;
| | - Lucia Catucci
- Department of Chemistry, University of Bari, Via Orabona 4, 70126 Bari, Italy;
| |
Collapse
|
20
|
Kesharwani P, Md S, Alhakamy NA, Hosny KM, Haque A. QbD Enabled Azacitidine Loaded Liposomal Nanoformulation and Its In Vitro Evaluation. Polymers (Basel) 2021; 13:250. [PMID: 33451016 PMCID: PMC7828524 DOI: 10.3390/polym13020250] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 12/13/2022] Open
Abstract
Azacitidine (AZA), an inhibitor of DNA methyltransferase, is a commonly recognized drug used in clinical treatment for myelodysplastic syndrome and breast cancer. Due to higher aqueous solubility and negative log P of AZA causes poor cancer cell permeation and controlled release. The objective of the present study was to formulate and optimize AZA-loaded liposome (AZA-LIPO) for breast cancer chemotherapy by using Box Behnken design (BBD) and in vitro evaluation using MCF-7 cells. AZA-LIPO were prepared using a thin film hydration technique and characterization study was performed by using FTIR and DSC. The prepared formulations were optimized using BBD and the optimized formulation was further subjected for particle size, surface charges, polydispersity index (PDI), drug loading, entrapment efficiency, TEM, XRD, in-vitro drug release and hemolytic toxicity. The mean particle size of optimized AZA-LIPO was 127 nm. Entrapment efficiency and drug loading of AZA-LIPO was found to be 85.2% ± 0.5 and 6.82 ± 1.6%, respectively. Further, in vitro drug release study showed preliminary burst release in 2 h followed by a sustained release for 36 h in phosphate buffer at different pH (4.0, 5.5, and 7.4) as compared to free drug. Drug release was found to be pH dependent, as the pH was increased, the drug release rate was found to be low. Time-dependent cell viability assay exhibited significant higher cell viability and higher internalization than free AZA in MCF-7 cells. AZA-LIPO were more effective than the free AZA in reducing Bcl2 expression, while increasing pro-apoptotic Bax and caspase-3 activity. The result showed that the formulated biocompatible AZA-LIPO nano-formulations may be used as an efficient anti-cancer drug delivery system for the treatment of breast cancer after establishing preclinical and clinical studies.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.M.); (N.A.A.); (K.M.H.)
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.M.); (N.A.A.); (K.M.H.)
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Khaled M. Hosny
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.M.); (N.A.A.); (K.M.H.)
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Anzarul Haque
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-kharj 16278, Saudi Arabia;
| |
Collapse
|
21
|
|