1
|
Salahuddin A, Ashraf A, Ahmad K, Hou H. Recent advances in chitosan-based smart hydrogel for drug delivery systems. Int J Biol Macromol 2024:135803. [PMID: 39419682 DOI: 10.1016/j.ijbiomac.2024.135803] [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: 06/11/2024] [Revised: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Achieving sustainable and controllable drug delivery is a highly effective disease treatment approach. Chitosan hydrogels, with their unique three-dimensional (3D) porous structures, offer tunable capacity, controllable degradation, various stimuli sensitivities, and the ability to encapsulate therapeutic agents. These characteristics provide chitosan hydrogels with inherent advantages as vehicles for drug delivery systems. In recent years, there has been a notable shift toward embracing the "back-to-nature" ethos, with biomass materials emerging as promising candidates for constructing chitosan hydrogels used in controlled drug release applications. This trend is sustained by their biodegradability, biocompatibility, and non-toxic properties, emphasizing their unique benefits and innovative features. These hydrogels exhibit sensitivity to various factors such as temperature, pH, ion concentration, light, magnetic fields, redox, ultrasound, and multi-responsiveness, offering opportunities for finely tuned drug release mechanisms. This review comprehensively outlines fabrication methods, properties, and biocompatibility of chitosan hydrogel, as well as modification strategies and stimuli-responsive mechanisms. Furthermore, their potential applications in subcutaneous (wound dressing), parental (transdermal drug delivery), oral (gastrointestinal tract), and facial (ophthalmic and brain) drug delivery are briefly discussed. The challenges in clinical application and the future outlook of chitosan-based smart hydrogel are also highlighted.
Collapse
Affiliation(s)
- Aiman Salahuddin
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No.1299, Sansha Road, Qingdao, Shandong Province 266404, PR China
| | - Azqa Ashraf
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No.1299, Sansha Road, Qingdao, Shandong Province 266404, PR China
| | - Khurshid Ahmad
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No.1299, Sansha Road, Qingdao, Shandong Province 266404, PR China
| | - Hu Hou
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No.1299, Sansha Road, Qingdao, Shandong Province 266404, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, Shandong Province 266237, PR China; Sanya Oceanographic Institution, Ocean University of China, Sanya, Hainan Province 572024, PR China; Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao, Shandong Province 266000, PR China.
| |
Collapse
|
2
|
Xia Y, Ma Z, Wu X, Wei H, Zhang H, Li G, Qian Y, Shahriari-Khalaji M, Hou K, Cao R, Zhu M. Advances in Stimuli-Responsive Chitosan Hydrogels for Drug Delivery Systems. Macromol Biosci 2024; 24:e2300399. [PMID: 38011585 DOI: 10.1002/mabi.202300399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/29/2023] [Indexed: 11/29/2023]
Abstract
Sustainable and controllable drug transport is one of the most efficient ways of disease treatment. Due to high biocompatibility, good biodegradability, and low costs, chitosan and its derivatives are widely used in biomedical fields. Specifically, chitosan hydrogel enables drugs to pass through biological barriers because of their abundant amino and hydroxyl groups that can interact with human tissues. Moreover, the multi-responsive nature (pH, temperature, ions strength, and magnetic field, etc.) of chitosan hydrogels makes precise drug release a possibility. Here, the synthesis methods, modification strategies, stimuli-responsive mechanisms of chitosan-based hydrogels, and their recent progress in drug delivery are summarized. Chitosan hydrogels that carry and release drugs through subcutaneous (dealing with wound dressing), oral (dealing with gastrointestinal tract), and facial (dealing with ophthalmic, ear, and brain) are reviewed. Finally, challenges toward clinic application and the future prospects of stimuli-responsive chitosan-based hydrogels are indicated.
Collapse
Affiliation(s)
- Yuhan Xia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Zhiyuan Ma
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Xuechen Wu
- Shanghai Starriver Bilingual School, Shanghai, 201108, China
| | - Huidan Wei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Han Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Guang Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Yuqi Qian
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Mina Shahriari-Khalaji
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Kai Hou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Ran Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, P. R. China
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
3
|
Myat YY, Sahatsapan N, Rojanarata T, Ngawhirunpat T, Opanasopit P, Pornpitchanarong C, Patrojanasophon P. Antibody-decorated chitosan-iodoacetamide-coated nanocarriers for the potential delivery of doxorubicin to breast cancer cells. Int J Biol Macromol 2024; 258:128797. [PMID: 38104687 DOI: 10.1016/j.ijbiomac.2023.128797] [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: 09/11/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
Using an active targeting approach of chemotherapeutics-loaded nanocarriers (NCs) with monoclonal antibodies is a potential strategy to improve the specificity of the delivery systems and reduce adverse reactions of chemotherapeutic drugs. Specific targeting of the human epidermal growth factor receptor-2 (HER-2), expressed excessively in HER-2-positive breast cancer cells, can be achieved by conjugating NCs with an anti-HER-2 monoclonal antibody. We constructed trastuzumab-conjugated chitosan iodoacetamide-coated NCs containing doxorubicin (Tras-Dox-CHI-IA-NCs) as a tumor-targeted drug delivery system, during the study. Chitosan-iodoacetamide (CHI-IA) was synthesized and utilized to prepare trastuzumab-conjugated NCs (Tras-NCs). The morphology, physicochemical properties, drug loading, drug release, and biological activities of the NCs were elucidated. The Tras-NCs were spherical, with a particle size of approximately 76 nm, and had a positive zeta potential; after incorporating the drug, the size of the Tras-NC increased. A prolonged, 24-h drug release from the NCs was achieved. The Tras-NCs exhibited high cellular accumulation and significantly higher antitumor activity against HER-2-positive breast cancer cells than the unconjugated NCs and the drug solution. Therefore, Tras-Dox-CHI-IA-NCs could be a promising nanocarrier for HER-2-positive breast cancer.
Collapse
Affiliation(s)
- Yin Yin Myat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Nitjawan Sahatsapan
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague 160 00, Czech Republic
| | - Theerasak Rojanarata
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Tanasait Ngawhirunpat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Chaiyakarn Pornpitchanarong
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| | - Prasopchai Patrojanasophon
- Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
4
|
Fan M, Huang Y, Zhu X, Zheng J, Du M. Octreotide and Octreotide-derived delivery systems. J Drug Target 2023; 31:569-584. [PMID: 37211679 DOI: 10.1080/1061186x.2023.2216895] [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: 02/15/2023] [Revised: 04/01/2023] [Accepted: 04/29/2023] [Indexed: 05/23/2023]
Abstract
Pharmaceutical peptide Octreotide is a somatostatin analog with targeting and therapeutic abilities. Over the last decades, Octreotide has been developed and approved to treat acromegaly and neuroendocrine tumours, and Octreotide-based radioactive conjugates have been leveraged clinically to detect small neuroendocrine tumour sites. Meanwhile, variety of Octreotide-derived delivery strategies have been proposed and explored for tumour targeted therapeutics or diagnostics in preclinical or clinical settings. In this review, we especially focus on the preclinical development and applications of Octreotide-derived drug delivery systems, diagnostic nanosystems, therapeutic nanosystems and multifunctional nanosystems, we also briefly discuss challenges and prospects of these Octreotide-derived delivery systems.
Collapse
Affiliation(s)
- Mingliang Fan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yue Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Xinlin Zhu
- Department of Dermatology, Shanghai Key Laboratory of Medical Mycology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jiayu Zheng
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Mingwei Du
- Department of Dermatology, Shanghai Key Laboratory of Medical Mycology, Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| |
Collapse
|
5
|
Sandal P, Kumari L, Patel P, Singh A, Singh D, Gupta GD, Kurmi BD. Doxorubicin Conjugates: An Efficient Approach for Enhanced Therapeutic Efficacy with Reduced Side Effects. Assay Drug Dev Technol 2023; 21:137-156. [PMID: 37083490 DOI: 10.1089/adt.2022.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023] Open
Abstract
Continuous drug delivery modification is the scientific approach and is a basic need for the efficient therapeutic efficacy of active drug molecules. Polymer-drug conjugates have long been a hallmark of the drug delivery sector, with various conjugates on the market or in clinical trials. Improved drug solubilization, extended blood circulation, decreased immunogenicity, controlled release behavior, and increased safety are the advantages of conjugating drugs to the polymeric carrier like polyethylene glycol (PEG). Polymer therapies have evolved over the last decade, resulting in polymer-drug conjugates with diverse topologies and chemical properties. Traditional nondegradable polymeric carriers like PEG and hydroxy propyl methacrylate have been clinically employed to fabricate polymer-drug conjugates. Still, functionalized polymer-drug conjugates are increasingly being used to increase localized drug delivery and ease of removal. Researchers have developed multifunctional carriers that can "see and treat" patients using medicinal and diagnostic chemicals. This review focused on the various conjugation approaches for attaching the doxorubicin to different polymers to achieve enhanced therapeutic efficacy, that is, increased bioavailability and reduced adverse effects.
Collapse
Affiliation(s)
- Pallavi Sandal
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | - Lakshmi Kumari
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Punjab, India
| | - Amrinder Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | - Dilpreet Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| | | | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, Punjab, India
| |
Collapse
|
6
|
Kantak MN, Bharate SS. Analysis of clinical trials on biomaterial and therapeutic applications of chitosan: A review. Carbohydr Polym 2022; 278:118999. [PMID: 34973801 DOI: 10.1016/j.carbpol.2021.118999] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023]
Abstract
Chitosan is a modified natural carbohydrate polymer derived from chitin that occurs in many natural sources. It has a diverse range of applications in medical and pharmaceutical sciences. Its primary and permitted use is biomaterial in medical devices. Chitosan and its derivatives also find utility in pharmaceuticals as an excipient, drug carrier, or therapeutic agent. The USFDA has approved chitosan usage as a biomaterial but not for pharmaceutical use, primarily because of the concerns over its source, purity, and immunogenicity. A large number of clinical studies are underway on chitosan-based materials/ products because of their diverse applications. Herein, we analyze clinical studies to understand their clinical usage portfolio. Our analysis shows that >100 clinical studies are underway to investigate the safety/efficacy of chitosan or its biomaterials/ nanoparticles, comprising ~95% interventional and ~ 5% observational studies. The regulatory considerations that limit the use of chitosan in pharmaceuticals are also deliberated. TEASER: Clinical Trials of Chitosan.
Collapse
Affiliation(s)
- Maithili N Kantak
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - Sonali S Bharate
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India.
| |
Collapse
|
7
|
Rana A, Bhatnagar S. Advancements in folate receptor targeting for anti-cancer therapy: A small molecule-drug conjugate approach. Bioorg Chem 2021; 112:104946. [PMID: 33989916 DOI: 10.1016/j.bioorg.2021.104946] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/17/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Targeted delivery combined with controlled release of drugs has a crucial role in future of personalized medicine. The majority of cancer drugs are intended to interfere with one or more cellular events. Anticancer agents can also be toxic to healthy cells, as healthy cells may also need to proliferate and avoid apoptosis. The focus of this review covers the principles, advantages, drawbacks and summarize criteria that must be met for design of small molecule-drug conjugates (SMDCs) to achieve the desired therapeutic potency with minimal toxicity. SMDCs are composed of a targeting ligand, a releasable bridge, a spacer, and a therapeutic payload. We summarize the criteria for the effective design that influences the selection of tumor specific receptor and optimum elements in the design of SMDCs. We also discuss the criteria for selecting the optimal therapeutic drug payload, spacer and linker. The linker chemistries and cleavage strategies are also discussed. Finally, we review the folate receptor targeting SMDCs that are in preclinical development and in clinical trials.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| |
Collapse
|
8
|
Rostami E. Progresses in targeted drug delivery systems using chitosan nanoparticles in cancer therapy: A mini-review. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101813] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
9
|
Kim H, Kwak G, Kim K, Yoon HY, Kwon IC. Theranostic designs of biomaterials for precision medicine in cancer therapy. Biomaterials 2019; 213:119207. [DOI: 10.1016/j.biomaterials.2019.05.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 02/06/2023]
|
10
|
Xu W, Wang H, Dong L, Zhang P, Mu Y, Cui X, Zhou J, Huo M, Yin T. Hyaluronic acid-decorated redox-sensitive chitosan micelles for tumor-specific intracellular delivery of gambogic acid. Int J Nanomedicine 2019; 14:4649-4666. [PMID: 31303753 PMCID: PMC6603291 DOI: 10.2147/ijn.s201110] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction: Herein, a hyaluronic acid (HA)-coated redox-sensitive chitosan-based nanoparticle, HA(HECS-ss-OA)/GA, was successfully developed for tumor-specific intracellular rapid delivery of gambogic acid (GA). Materials and methods: The redox-sensitive polymer, HECS-ss-OA, was prepared through a well-controlled synthesis procedure with a satisfactory reproducibility and stable resulted surface properties of the assembled cationic micelles. GA was solubilized into the inner core of HECS-ss-OA micelles, while HA was employed to coat outside HECS-ss-OA/GA for CD44-mediated active targeting along with protection from cation-associated in vivo defects. The desirable redox-sensitivity of HA(HECS-ss-OA)/GA was demonstrated by morphology and particle size changes alongside in vitro drug release of nanoparticles in different simulated reducing environments. Results: The results of flow cytometry and confocal microscopy confirmed the HA-receptor mediated cellular uptake and burst drug release in highly reducing cytosol of HA(HECS-ss-OA)/GA. Consequently, HA(HECS-ss-OA)/GA showed the highest apoptosis induction and cytotoxicity over the non-sensitive (HA(HECS-cc-OA)/GA) and HA un-coated (HECS-ss-OA/GA) controls against A549 NSCLC model both in vitro and in vivo. Furthermore, a diminished systemic cytotoxicity was observed in HA(HECS-ss-OA)/GA treated mice compared with those treated by HA un-coated cationic ones and GA solution.
Collapse
Affiliation(s)
- Wei Xu
- Department of Pharmacy, Shandong Provincial Qian Foshan Hospital, Shandong University, Jinan 250014, People's Republic of China.,Qianfoshan Hospital, The First Hospital Affiliation with Shandong First Medical University, Jinan 250012, People's Republic of China
| | - Honglan Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lihui Dong
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Pan Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yan Mu
- Qianfoshan Hospital, The First Hospital Affiliation with Shandong First Medical University, Jinan 250012, People's Republic of China
| | - Xueyan Cui
- Qianfoshan Hospital, The First Hospital Affiliation with Shandong First Medical University, Jinan 250012, People's Republic of China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Meirong Huo
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Tingjie Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| |
Collapse
|
11
|
Zhang E, Xing R, Liu S, Qin Y, Li K, Li P. Advances in chitosan-based nanoparticles for oncotherapy. Carbohydr Polym 2019; 222:115004. [PMID: 31320066 DOI: 10.1016/j.carbpol.2019.115004] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/27/2019] [Accepted: 06/17/2019] [Indexed: 01/02/2023]
Abstract
Chitosan has attracted considerable attention as an anti-tumor drug carrier material in recent years, which is due to its biocompatibility and biodegradability, as well as the simple and mild preparing techniques of drug-loaded nanoparticles. Chitosan-based nanoparticles can deliver various anti-tumor agents to specific tumor tissues by passive and active targeting mechanisms, including traditional chemotherapeutic agents, DNA or siRNA, proteins, photosensitizers and so on. In this review, we summarized the factors affecting the anti-tumor efficacy of chitosan-based nanoparticles, to aid exploring the function-structure relationship. The recent studies on chitosan-based nanoparticles for oncotherapy were highlighted, including their structures, properties and pharmacological effects. Finally, we offered our perspectives on the challenges and future development of this area.
Collapse
Affiliation(s)
- Enhui Zhang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Ronge Xing
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
| | - Song Liu
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Yukun Qin
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Kecheng Li
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| | - Pengcheng Li
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China.
| |
Collapse
|
12
|
Xie D, Du J, Bao M, Zhou A, Tian C, Xue L, Ju C, Shen J, Zhang C. A one-pot modular assembly strategy for triple-play enhanced cytosolic siRNA delivery. Biomater Sci 2019; 7:901-913. [PMID: 30575823 DOI: 10.1039/c8bm01454j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Robust efficiency for cytosolic small interfering RNA (siRNA) delivery is of great importance for effective gene therapy. To significantly improve the cytosolic siRNA delivery, a "one-pot modular assembly" strategy is developed to assemble a triple-play enhanced cytosolic siRNA delivery system via a facile and innocuous copper-free click reaction. Specifically, three modules are prepared including octreotide for receptor-mediated endocytosis, a cell-penetrating peptide (CPP) for cell penetration, and glutamic acid for the charge-reversal property. All three modules with distinct facilitating endocytosis effects are expediently assembled on the surface of the siRNA/liposome complex to fabricate a multifunctional integrated siRNA delivery system (OCA-CC). OCA-CC has been demonstrated to have enhanced cytosolic delivery and superior gene-silencing efficiency in multiple tumor cells due to the combined effects of all the three modules. High levels of survivin-silencing are also achieved by OCA-CC on orthotopic human breast cancer (MCF-7)-bearing mice accompanied by significant tumor inhibition. This research provides a facile strategy to produce safe and tunable siRNA delivery systems for effective gene therapy and to facilitate the development of multifunctional siRNA vectors.
Collapse
Affiliation(s)
- Daping Xie
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Ren SZ, Zhu D, Zhu XH, Wang B, Yang YS, Sun WX, Wang XM, Lv PC, Wang ZC, Zhu HL. Nanoscale Metal-Organic-Frameworks Coated by Biodegradable Organosilica for pH and Redox Dual Responsive Drug Release and High-Performance Anticancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:20678-20688. [PMID: 31081332 DOI: 10.1021/acsami.9b04236] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Responsive nanocarriers with biocompatibility and precise drug releasing capability have emerged as a prospective candidate for anticancer treatment. However, the challenges imposed by the complicated preparation process and limited loading capacities have seriously impeded the development of novel multifunctional drug delivery systems. Here, we developed a novel and dual-responsive nanocarrier based on a nanoscale ZIF-8 core and an organosilica shell containing disulfide bridges in its frameworks through a facile and efficient strategy. The prepared ZIF-8@DOX@organosilica nanoparticles (ZDOS NPs) exhibited a well-defined structure and excellent doxorubicin (DOX) loading capability (41.2%) with pH and redox dual-sensitive release properties. The degradation of the organosilica shell was observed after 12 h incubation with a 10 mM reducing agent. Confocal imaging and flow cytometry analysis further proved that the nanocarriers can efficiently enter cells and complete intracellular DOX release under the low pH and high glutathione concentrations, which resulted in an enhanced cytotoxicity of DOX for cancer cells. Meanwhile, subcellular localization experiments revealed that the ZDOS NPs entered cells mainly by endocytosis and then escaped from lysosomes into the cytosol. Moreover, in vivo assays also demonstrated that the ZDOS NPs exhibited negligible systemic toxicity and significantly enhanced anticancer efficiencies compared with free DOX. In summary, our prepared pH and redox dual-responsive nanocarriers provide a potential platform for controlled release and cancer treatment.
Collapse
|
14
|
Wang T, Cai ZS, Zhang TT, Li M, Fang GG, Zhu XM. Dehydroabietyl Glycidyl Ether Grafted Hydroxyethyl Chitosan: Synthesis, Characterization and Physicochemical Properties. TENSIDE SURFACT DET 2019. [DOI: 10.3139/113.110618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AbstractA series of novel polymeric nonionic surfactants based on water-soluble N,O-hydroxyethyl chitosan (N,O-HECTS) and dehydroabietyl glycidyl ether (DAGE), DAGE-g-N,O-HECTSs, were synthesized by an additive reaction between N,O-HECTS and DAGE. The structures of DAGE-g-N,O-HECTSs were characterized by FT-IR and 1H NMR. The substitution degree of hydroxyethylation (DSHE) of N,O-HECTS and the grafting degree (DG) of DAGE onto N,O-HECTS for DAGE-g-N,O-HECTSs were determined by elemental analysis. The surface activities of DAGE-g-N,O-HECTSs in aqueous solution were investigated by measuring the surface tension. The experimental results showed that the degree of grafting (DG) of DAGE-gN,O-HECTSs could have a significant impact on their critical micelle concentrations (CMCs) and surface tensions at the CMC (γCMC), but the DG of DAGE-g-N,O-HECTSs had almost no effect on the minimum of surface tensions (γmin). When using the DAGE-g-N,O-HECTSs as emulsifier, the increase in DG had a favorable influence on the stability of an emulsion of water and benzene. At a DG greater than 40.45%, the emulsifying power of DAGE-g-N,O-HECTS exceeded that of Tween-60.
Collapse
Affiliation(s)
- Ting Wang
- 1School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng 224051 Jiangsu Province, P.R. China
| | - Zhao-sheng Cai
- 1School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng 224051 Jiangsu Province, P.R. China
| | - Ting-ting Zhang
- 1School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng 224051 Jiangsu Province, P.R. China
| | - Man Li
- 1School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng 224051 Jiangsu Province, P.R. China
- 2Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042 Jiangsu Province, P.R China
| | - Gui-gan Fang
- 2Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042 Jiangsu Province, P.R China
| | - Xue-mei Zhu
- 1School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng 224051 Jiangsu Province, P.R. China
| |
Collapse
|
15
|
Lu A, Wang J, Najarro MC, Li S, Deratani A. Synthesis and self-assembly of AB2-type amphiphilic copolymers from biobased hydroxypropyl methyl cellulose and poly(L-lactide). Carbohydr Polym 2019; 211:133-140. [DOI: 10.1016/j.carbpol.2019.01.110] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/28/2019] [Accepted: 01/31/2019] [Indexed: 01/22/2023]
|
16
|
Wu S, Bin W, Tu B, Li X, Wang W, Liao S, Sun C. A Delivery System for Oral Administration of Proteins/Peptides Through Bile Acid Transport Channels. J Pharm Sci 2019; 108:2143-2152. [PMID: 30721709 DOI: 10.1016/j.xphs.2019.01.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/13/2019] [Accepted: 01/28/2019] [Indexed: 12/15/2022]
Abstract
Proteins and peptides are poorly absorbed via oral administration because of the gastrointestinal tract environment and lysosomal digestion after apical endocytosis. A delivery system, consisting of a deoxycholic acid-conjugated nanometer-sized carrier, may enhance the absorption of proteins in the intestine via the bile acid pathway. Deoxycholic acid is first conjugated to chitosan. Liposomes are then prepared and loaded with the model drug insulin. Finally, the conjugates are bound to the liposome surface to form deoxycholic acid and chitosan conjugate-modified liposomes (DC-LIPs). This study demonstrates that DC-LIPs can promote the intestinal absorption of insulin via the apical sodium-dependent bile acid transporter, based on observing fluorescently stained tissue slices of the rat small intestine and a Caco-2 cell uptake experiment. Images of intestinal slices revealed that excellent absorption of DC-LIPs is achieved via apical sodium-dependent bile acid transporter, and a flow cytometry experiment proved that DC-LIPs are a highly efficient delivery carrier. Caco-2 cells were also used to study the lysosome escape ability of DC-LIPs. We learned from confocal microscopy photographs that DC-LIPs can protect their contents from being destroyed by the lysosome. Finally, according to pharmacokinetic analyses, insulin-loaded DC-LIPs show a significant hypoglycemic effect with an oral bioavailability of 16.1% in rats with type I diabetes.
Collapse
Affiliation(s)
- Siwen Wu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Wen Bin
- Department of Biochemistry and Molecular Biology, School of Life Sciences and Biological Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Biyun Tu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Xifeng Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Wei Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Suling Liao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Changshan Sun
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
17
|
Anticancer effect of intracellular-delivered paclitaxel using novel pH-sensitive LMWSC-PCL di-block copolymer micelles. J IND ENG CHEM 2019. [DOI: 10.1016/j.jiec.2018.09.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
18
|
Abdellatif AA, Aldalaen SM, Faisal W, Tawfeek HM. Somatostatin receptors as a new active targeting sites for nanoparticles. Saudi Pharm J 2018; 26:1051-1059. [PMID: 30416362 PMCID: PMC6218373 DOI: 10.1016/j.jsps.2018.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
The delivery of nanoparticles through receptor-mediated cell interactions has nowadays a major attention in the area of drug targeting applications. This specific kind of targeting is mediated by localized receptors impeded into the target site with subsequent drugs internalization. Hence, this type of interaction would diminish side effects and enhance drug delivery efficacy to the target site. Somatostatin receptors (SSTRs) are one type of G protein-coupled receptors, which could be active targeted for various purposes. There are five SSTRs types (SSTR1-5) which are localized at various organs in the body and spread into different tissues. SSTRs could be considered as a promising target to various nanoparticles which is facilitated when nanoparticles are modified through specific ligand or coating to allow better binding. This review discusses the exploration of SSTRs for active targeting of nanoparticles with certain emphasize on their interaction at the cellular level.
Collapse
Affiliation(s)
- Ahmed A.H. Abdellatif
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy, Qassim University, Buraydah, 51452 Al-Qassim, Kingdom of Saudi Arabia
| | - Sa'ed M. Aldalaen
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Mutah University, Mutah, Al-Karak 61710, Jordan
| | - Waleed Faisal
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt
- School of Pharmacy, University of College Cork, Cork, Ireland
| | - Hesham M. Tawfeek
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Mutah University, Mutah, Al-Karak 61710, Jordan
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| |
Collapse
|
19
|
Li L, Wang Q, Zhang X, Luo L, He Y, Zhu R, Gao D. Dual-targeting liposomes for enhanced anticancer effect in somatostatin receptor II-positive tumor model. Nanomedicine (Lond) 2018; 13:2155-2169. [DOI: 10.2217/nnm-2018-0115] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: We developed octreotide-modified magnetic liposomes (OMlips) as dual-targeting drug carriers to enhance the drug accumulation in tumor site. Materials & methods: Octreotide acts as a modified ligand for receptor-mediated targeting and the coated Fe3O4 nanoparticles offer the magnetic targeting property. SSTR2 overexpressed A549 cells and S180 cells were chosen to explore the targeting ability and antitumor effect of the oleanolic acid (OA)-loaded OMlips in vitro and in vivo. Results: The OMlips platform significantly improves the targeting, penetrating and accumulation of OA at the SSTR2 overexpressed cells and SSTR2-positive tumor-bearing mice. Conclusion: The OA-loaded OMlips have better antitumor effect and lower systemic toxicity. Such a receptor-mediated and magnetically-orienting dual-targeting drug nanocarriers may have great potentials in clinical practice.
Collapse
Affiliation(s)
- Lei Li
- Applying Chemistry Key Lab of Hebei Province, Department of Bioengineer, Yanshan University, No.438 Hebei Street, Qinhuangdao, 066004, PR China
- State Key Laboratory of Metastable Materials Science & Technology, Yanshan University, No.438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Qianqian Wang
- Applying Chemistry Key Lab of Hebei Province, Department of Bioengineer, Yanshan University, No.438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Xuwu Zhang
- Applying Chemistry Key Lab of Hebei Province, Department of Bioengineer, Yanshan University, No.438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Liyao Luo
- Applying Chemistry Key Lab of Hebei Province, Department of Bioengineer, Yanshan University, No.438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Yuchu He
- Applying Chemistry Key Lab of Hebei Province, Department of Bioengineer, Yanshan University, No.438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Ruiyan Zhu
- Applying Chemistry Key Lab of Hebei Province, Department of Bioengineer, Yanshan University, No.438 Hebei Street, Qinhuangdao, 066004, PR China
- Hebei Province Asparagus Industry Technology Research Institute, No.12 Donghai Road, Qinhuangdao, 066318, PR China
| | - Dawei Gao
- Applying Chemistry Key Lab of Hebei Province, Department of Bioengineer, Yanshan University, No.438 Hebei Street, Qinhuangdao, 066004, PR China
- State Key Laboratory of Metastable Materials Science & Technology, Yanshan University, No.438 Hebei Street, Qinhuangdao, 066004, PR China
- Hebei Province Asparagus Industry Technology Research Institute, No.12 Donghai Road, Qinhuangdao, 066318, PR China
| |
Collapse
|
20
|
Paramonov VM, Desai D, Kettiger H, Mamaeva V, Rosenholm JM, Sahlgren C, Rivero-Müller A. Targeting Somatostatin Receptors By Functionalized Mesoporous Silica Nanoparticles - Are We Striking Home? Nanotheranostics 2018; 2:320-346. [PMID: 30148051 PMCID: PMC6107779 DOI: 10.7150/ntno.23826] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 06/25/2018] [Indexed: 12/02/2022] Open
Abstract
The concept of delivering nanoformulations to desired tissues by means of targeting membrane receptors of high local abundance by ligands anchored to the nanocarrier has gained a lot of attention over the last decade. Currently, there is no unanimous opinion on whether surface functionalization of nanocarriers by targeting ligands translates into any real benefit in terms of pharmacokinetics or treatment outcomes. Having examined the published nanocarriers designed to engage with somatostatin receptors, we realized that in the majority of cases targetability claims were not supported by solid evidence of targeting ligand-targeted receptor coupling, which is the very crux of a targetability concept. Here, we present an approach to characterize targetability of mesoporous silica-based nanocarriers functionalized with ligands of somatostatin receptors. The targetability proof in our case comes from a functional assay based on a genetically-encoded cAMP probe, which allows for real-time capture of receptor activation in living cells, triggered by targeting ligands on nanoparticles. We elaborate on the development and validation of the assay, highlighting the power of proper functional tests in the characterization pipeline of targeted nanoformulations.
Collapse
Affiliation(s)
- Valeriy M Paramonov
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Diti Desai
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Helene Kettiger
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Veronika Mamaeva
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Cecilia Sahlgren
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Adolfo Rivero-Müller
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Poland
| |
Collapse
|
21
|
Poudel AJ, He F, Huang L, Xiao L, Yang G. Supramolecular hydrogels based on poly (ethylene glycol)-poly (lactic acid) block copolymer micelles and α-cyclodextrin for potential injectable drug delivery system. Carbohydr Polym 2018; 194:69-79. [DOI: 10.1016/j.carbpol.2018.04.035] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/16/2018] [Accepted: 04/07/2018] [Indexed: 01/15/2023]
|
22
|
Zhang J, Wang Y, Jiang Y, Liu T, Luo Y, Diao E, Cao Y, Chen L, Zhang L, Gu Q, Zhou J, Sun F, Zheng W, Liu J, Li X, Hu W. Enhanced cytotoxic and apoptotic potential in hepatic carcinoma cells of chitosan nanoparticles loaded with ginsenoside compound K. Carbohydr Polym 2018; 198:537-545. [PMID: 30093032 DOI: 10.1016/j.carbpol.2018.06.121] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 06/26/2018] [Accepted: 06/28/2018] [Indexed: 11/18/2022]
Abstract
Ginsenoside compound K (CK) has been shown to exhibit anticancer properties. In this study, chitosan nanoparticles loaded with ginsenoside compound K (CK-NPs) were prepared as a delivery system using a self-assembly technique with amphipathic deoxycholic acid-O carboxymethyl chitosan as the carrier, which improved the water solubility of CK. By evaluating drug loading, entrapment efficiency, and in vitro release behavior, the feasibility of CK-NPs as a drug carrier nanoparticle for the treatment of human hepatic carcinoma cells (HepG2) was investigated. Result revealed that CK and CK-NPs showed a dose-dependent inhibitory effect on HepG2 cells with IC50 values of 23.33 and 16.58 μg/mL, respectively. Furthermore, fluorescence imaging demonstrated that CK-NPs promoted cellular uptake in vitro. Therefore, all results indicated that CK-NPs might be a novel drug delivery system to improve the solubility and enhance the cytotoxic and apoptotic potentials of CK for effective liver cancer chemotherapy.
Collapse
Affiliation(s)
- Jianmei Zhang
- College of Food Science and Pharmacology, Xinjiang Agricultural University, Xinjiang, 830052, China; Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Yijun Wang
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Yunyao Jiang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Tingwu Liu
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Yanyan Luo
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Enjie Diao
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Yufeng Cao
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Liang Chen
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Liang Zhang
- College of Food Science and Pharmacology, Xinjiang Agricultural University, Xinjiang, 830052, China; Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Qian Gu
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Jinyi Zhou
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Fengting Sun
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Wancai Zheng
- College of Food Science and Pharmacology, Xinjiang Agricultural University, Xinjiang, 830052, China; Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China
| | - Jianxun Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Xueqin Li
- Department of Gerontology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huanghe West Road, Huaian, 223300, China.
| | - Weicheng Hu
- College of Food Science and Pharmacology, Xinjiang Agricultural University, Xinjiang, 830052, China; Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection/Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China.
| |
Collapse
|
23
|
Wang X, Gu X, Wang H, Yang J, Mao S. Enhanced delivery of doxorubicin to the liver through self-assembled nanoparticles formed via conjugation of glycyrrhetinic acid to the hydroxyl group of hyaluronic acid. Carbohydr Polym 2018; 195:170-179. [PMID: 29804965 DOI: 10.1016/j.carbpol.2018.04.052] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/13/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022]
Abstract
Liver-targeted nanoparticles is highly desired for better therapy of liver cancer. In this study, enhanced delivery of doxorubicin (DOX) to the liver cells through self-assembled nanoparticles formed via conjugation of glycyrrhetinic acid (GA) to the hydroxyl group of hyaluronic acid (HA) was investigated. The DOX loaded hyaluronic acid-glycyrrhetinic acid succinate (HSG) conjugates based nanoparticles (HSG/DOX nanoparticles) were sub-spherical in shape with particle size in the range of 180-280 nm, the drug loading was drug-to-carrier ratio and GA graft ratio dependent. In vitro release study suggested that the release of DOX from HSG nanoparticles was sustained and the release rate was pH and GA graft ratio dependent. MTT assay indicated the HSG/DOX nanoparticles presented a GA-dependent cytotoxicity to HepG2 cells. Pharmacokinetics study demonstrated the HSG/DOX nanoparticles could prolong blood circulation time of DOX and had a higher AUC value than that of DOX solution. Furthermore, tissue distribution study revealed the HSG/DOX nanoparticles significantly increased the accumulation of DOX in the liver and meanwhile decreased the cardiotoxicity and nephrotoxicity of DOX. Moreover, the liver targeting enhancing capacity was HSG conjugate structure dependent. The accumulation of HSG-20/DOX, HSG-12/DOX, and HSG-6/DOX nanoparticles in the liver was 4.0-, 3.1-, and 2.6-fold higher than that of DOX solution. In vivo imaging analysis further demonstrated HSG nanoparticles not only had better liver targeting effect, but also presented superior tumor targeting efficiency, and the tumor targeting capacity was also GA-dependent. These results indicated that HSG conjugates prepared via modifying the hydroxyl groups of HA have promising potential as a liver-targeting nanocarrier for the delivery of hydrophobic anti-tumor drugs.
Collapse
Affiliation(s)
- Xiaodan Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiangqin Gu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huimin Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
24
|
Farhangi M, Kobarfard F, Mahboubi A, Vatanara A, Mortazavi SA. Preparation of an optimized ciprofloxacin-loaded chitosan nanomicelle with enhanced antibacterial activity. Drug Dev Ind Pharm 2018; 44:1273-1284. [PMID: 29452500 DOI: 10.1080/03639045.2018.1442847] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE The objective of this study was to evaluate the effect of lipid structure on physicochemical properties of chitosan-fatty acid nanomicelles and prepare an optimum ciprofloxacin-loaded formulation from these conjugates which could enhance the antibacterial effects of drug against some important pathogens like P. aeruginosa. SIGNIFICANCE Nowadays, resistance in infectious diseases is a growing worldwide concern. Nanocarriers can increase the therapeutic index and consequently reduce the antibiotic resistance. By site-specific delivery of drug, the adverse effects of broad-spectrum antibiotics such as ciprofloxacin would be reduced. METHODS Fatty acid grafted chitosan conjugates were synthetized in the presence of 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide. The effects of fatty acid type (stearic acid, palmitic acid, and linoleic acid) on physicochemical properties of conjugates were investigated. Ciprofloxacin was encapsulated in nanomicelles by thin film hydration method. Also, the preparation process was optimized with a central composite design. The antibacterial effect of optimum formulation against P. aeruginosa, K. pneumoniae, and S. pneumoniae species was determined. RESULTS All conjugates were synthetized with high yield values and the substitution degrees ranged between 2.13 and 35.46%. Ciprofloxacin was successfully encapsulated in nanomicelles. The optimum formulation showed high drug loading (≈ 19%), with particle size of about 260 nm and a sustained release profile of ciprofloxacin. The minimum inhibitory concentrations of ciprofloxacin in optimum formulation against P. aeruginosa and K. pneumoniae species were 4 and 2 times lower in comparison with the free drug, respectively. CONCLUSIONS The antibacterial effect of ciprofloxacin was improved by encapsulation of drug in chitosan nanomicelles.
Collapse
Affiliation(s)
- Mahdieh Farhangi
- a Department of Pharmaceutics, School of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Farzad Kobarfard
- b Department of Medicinal Chemistry, School of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Arash Mahboubi
- a Department of Pharmaceutics, School of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran.,c Food Safety Research Center, School of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Alireza Vatanara
- d Department of Pharmaceutics, Faculty of Pharmacy , Tehran University of Medical Sciences , Tehran , Iran
| | - Seyed Alireza Mortazavi
- a Department of Pharmaceutics, School of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
25
|
Zhang X, Xu G, Gadora K, Cheng H, Peng J, Ma Y, Guo Y, Chi C, Zhou J, Ding Y. Dual-sensitive chitosan derivative micelles for site-specific drug release in the treatment of chicken coccidiosis. RSC Adv 2018; 8:14515-14526. [PMID: 35540782 PMCID: PMC9079931 DOI: 10.1039/c8ra02144a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 04/02/2018] [Indexed: 12/17/2022] Open
Abstract
Here, we report a “dual-sensitive” drug delivery platform packaged with anti-coccidia drug diclazuril (DIC) applied in the field of intestinal-targeted administration.
Collapse
|
26
|
Deshmukh AS, Chauhan PN, Noolvi MN, Chaturvedi K, Ganguly K, Shukla SS, Nadagouda MN, Aminabhavi TM. Polymeric micelles: Basic research to clinical practice. Int J Pharm 2017; 532:249-268. [PMID: 28882486 DOI: 10.1016/j.ijpharm.2017.09.005] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/02/2017] [Accepted: 09/02/2017] [Indexed: 12/17/2022]
Abstract
Rapidly developing polymeric micelles as potential targeting carriers has intensified the need for better understanding of the underlying principles related to the selection of suitable delivery materials for designing, characterizing, drug loading, improving stability, targetability, biosafety and efficacy. The emergence of advanced analytical tools such as fluorescence resonance energy transfer and dissipative particle dynamics has identified new dimensions of these nanostructures and their behavior in much greater details. This review summarizes recent efforts in the development of polymeric micelles with respect to their architecture, formulation strategy and targeting possibilities along with their preclinical and clinical aspects. Literature of the past decade is discussed critically with special reference to the chemistry involved in the formation and clinical applications of these versatile materials. Thus, our main objective is to provide a timely update on the current status of polymeric micelles highlighting their applications and the important parameters that have led to successful delivery of drugs to the site of action.
Collapse
Affiliation(s)
- Anand S Deshmukh
- Department of Pharmaceutical Research, Shree Dhanvantary Pharmacy College, Kim, Surat, Gujarat 394 110, India.
| | - Pratik N Chauhan
- Department of Pharmaceutical Research, Shree Dhanvantary Pharmacy College, Kim, Surat, Gujarat 394 110, India
| | - Malleshappa N Noolvi
- Department of Pharmaceutical Research, Shree Dhanvantary Pharmacy College, Kim, Surat, Gujarat 394 110, India
| | - Kiran Chaturvedi
- Department of Pharmaceutical Research, Shree Dhanvantary Pharmacy College, Kim, Surat, Gujarat 394 110, India
| | - Kuntal Ganguly
- Department of Pharmaceutical Research, Shree Dhanvantary Pharmacy College, Kim, Surat, Gujarat 394 110, India
| | - Shyam S Shukla
- Department of Pharmaceutical Research, Shree Dhanvantary Pharmacy College, Kim, Surat, Gujarat 394 110, India
| | - Mallikarjuna N Nadagouda
- Department of Pharmaceutical Research, Shree Dhanvantary Pharmacy College, Kim, Surat, Gujarat 394 110, India
| | - Tejraj M Aminabhavi
- Department of Pharmaceutical Research, Shree Dhanvantary Pharmacy College, Kim, Surat, Gujarat 394 110, India.
| |
Collapse
|
27
|
Xiao L, Huang L, Moingeon F, Gauthier M, Yang G. pH-Responsive Poly(Ethylene Glycol)-block-Polylactide Micelles for Tumor-Targeted Drug Delivery. Biomacromolecules 2017; 18:2711-2722. [DOI: 10.1021/acs.biomac.7b00509] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Lin Xiao
- Department
of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lixia Huang
- Department
of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Firmin Moingeon
- Department
of Chemistry, University of Waterloo, Waterloo N2L 3G1, Canada
| | - Mario Gauthier
- Department
of Chemistry, University of Waterloo, Waterloo N2L 3G1, Canada
| | - Guang Yang
- Department
of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
28
|
Liu H, Wu S, Yu J, Fan D, Ren J, Zhang L, Zhao J. Reduction-sensitive micelles self-assembled from amphiphilic chondroitin sulfate A-deoxycholic acid conjugate for triggered release of doxorubicin. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 75:55-63. [PMID: 28415498 DOI: 10.1016/j.msec.2017.02.030] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/30/2016] [Accepted: 02/07/2017] [Indexed: 11/17/2022]
|
29
|
Jaskula–Sztul R, Chen G, Dammalapati A, Harrison A, Tang W, Gong S, Chen H. AB3-Loaded and Tumor-Targeted Unimolecular Micelles for Medullary Thyroid Cancer Treatment. J Mater Chem B 2017; 5:151-159. [PMID: 28025618 PMCID: PMC5180596 DOI: 10.1039/c6tb02530g] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Medullary thyroid cancer (MTC) is often resistant to standard therapies, emphasizing the need for the development of other treatments. A new histone deacetylase inhibitor, AB3, can effectively inhibit MTC cell proliferation in vitro. However, its poor aqueous solubility and stability, fast clearance, and lack of tumor targeting ability limit its in vivo application. Therefore, multifunctional unimolecular micelles were developed for targeted delivery of AB3 for MTC therapy. The unimolecular micelles exhibited a spherical core-shell structure, uniform size distribution, and excellent stability. AB3 was encapsulated into the hydrophobic core of the unimolecular micelles, thus significantly enhancing its aqueous solubility and stability. KE108, a somatostatin analog possessing high affinity to all five subtypes of SSTR, was used as an MTC-targeting ligand. In vitro cellular uptake analyses demonstrated that the KE108 exhibited superior targeting ability in MTC cells compared to octreotide, the first clinically used somatostatin analog. Moreover, the AB3-loaded and KE108-conjugated unimolecular micelles exhibited the best efficacy in suppressing MTC cell growth and tumor marker expression in vitro. Furthermore, AB3-loaded, KE108-conjugated micelles demonstrated the best anticancer efficacy in vivo without any apparent systemic toxicity, thereby offering a promising approach for targeted MTC therapy.
Collapse
Affiliation(s)
- Renata Jaskula–Sztul
- Department of Surgery, School of Medicine University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Guojun Chen
- Department of Materials Science and Engineering, University of Wisconsin–Madison, Madison, WI 53715, USA
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI 53715, USA
| | - Ajitha Dammalapati
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - April Harrison
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Weiping Tang
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Shaoqin Gong
- Department of Materials Science and Engineering, University of Wisconsin–Madison, Madison, WI 53715, USA
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI 53715, USA
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Herbert Chen
- Department of Surgery, School of Medicine University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
30
|
Wang X, Gu X, Wang H, Sun Y, Wu H, Mao S. Synthesis, characterization and liver targeting evaluation of self-assembled hyaluronic acid nanoparticles functionalized with glycyrrhetinic acid. Eur J Pharm Sci 2017; 96:255-262. [DOI: 10.1016/j.ejps.2016.09.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/08/2016] [Accepted: 09/26/2016] [Indexed: 01/16/2023]
|
31
|
Zhu X, Zhang Y, Huang H, Zhang H, Hou L, Zhang Z. Folic acid-modified and functionalized CuS nanocrystal-based nanoparticles for combined tumor chemo- and photothermal therapy. J Drug Target 2016; 25:425-435. [DOI: 10.1080/1061186x.2016.1266651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Xiali Zhu
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, Zhengzhou, PR China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Yingjie Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Heqing Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Huijuan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, PR China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou, PR China
| |
Collapse
|
32
|
Dehghankelishadi P, Saadat E, Ravar F, Safavi M, Pordeli M, Gholami M, Dorkoosh FA. In vitro and in vivo evaluation of paclitaxel–lapatinib-loaded F127 pluronic micelles. Drug Dev Ind Pharm 2016; 43:390-398. [DOI: 10.1080/03639045.2016.1254238] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Pouya Dehghankelishadi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterials Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Saadat
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ravar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Mahboobeh Pordeli
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mehdi Gholami
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Abedin Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterials Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Zhang X, Yang C, Zhou J, Huo M. Somatostatin Receptor-Mediated Tumor-Targeting Nanocarriers Based on Octreotide-PEG Conjugated Nanographene Oxide for Combined Chemo and Photothermal Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:3578-90. [PMID: 27244649 DOI: 10.1002/smll.201600618] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/18/2016] [Indexed: 05/28/2023]
Abstract
Nano-sized in vivo active targeting drug delivery systems have been developed to a high anti-tumor efficacy strategy against certain cancer-cells-specific. Graphene based nanocarriers with unique physical and chemical properties have shown significant potentials in this aspect. Here, octreotide (OCT), an efficient biotarget molecule, is conjugated to PEGylated nanographene oxide (NGO) drug carriers for the first time. The obtained NGO-PEG-OCT complex shows low toxicity and excellent stability in vivo and is able to achieve somatostatin receptor-mediated tumor-specific targeting delivery. Owing to the high loading efficiency and accurate targeting delivery of anti-cancer drug doxorubicin (DOX), our DOX loaded NGO-PEG-OCT complex offers a remarkably improved cancer-cell-specific cellular uptake, chemo-cytotoxicity, and decreased systemic toxicity compared to free DOX or NGO-PEG. More importantly, due to its strong near-infrared absorption, the NGO-PEG-OCT complex further enhances efficient photothermal ablation of tumors, delivering combined chemo and photothermal therapeutic effect against cancer cells.
Collapse
Affiliation(s)
- Xuyuan Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, China
| | - Chongyin Yang
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, China
| | - Meirong Huo
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, China
| |
Collapse
|
34
|
Basumallick S, Gabriela Nogueira Campos M, Richardson D, Gesquiere A, Santra S. Hydrothermally treated chitosan spontaneously forms water-soluble spherical particles stable at a wide pH range. INT J POLYM MATER PO 2016. [DOI: 10.1080/00914037.2016.1163568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
35
|
Liu T, Jia T, Yuan X, Liu C, Sun J, Ni Z, Xu J, Wang X, Yuan Y. Development of octreotide-conjugated polymeric prodrug of bufalin for targeted delivery to somatostatin receptor 2 overexpressing breast cancer in vitro and in vivo. Int J Nanomedicine 2016; 11:2235-50. [PMID: 27284243 PMCID: PMC4883818 DOI: 10.2147/ijn.s100404] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Development of polymeric prodrugs of small molecular anticancer drugs has become one of the most promising strategies to overcome the intrinsic shortcomings of small molecular anticancer drugs and improve their anticancer performance. MATERIALS AND METHODS In the current work, we fabricated a novel octreotide (Oct)-modified esterase-sensitive tumor-targeting polymeric prodrug of bufalin (BUF) and explored its anticancer performance against somatostatin receptor 2 overexpressing breast cancer. RESULTS The obtained tumor-targeting polymeric prodrug of BUF, P(oligo[ethylene glycol] monomethyl ether methacrylate [OEGMA]-co-BUF-co-Oct), showed a nanosize dimension and controlled drug release features in the presence of esterase. It was demonstrated by in vitro experiment that P(OEGMA-co-BUF-co-Oct) showed enhanced cytotoxicity, cellular uptake, and apoptosis in comparison with those of free BUF. In vivo experiment further revealed the improved accumulation of drugs in tumor tissues and enhanced anticancer performance of P(OEGMA-co-BUF-co-Oct). CONCLUSION Taken together, this study indicated that polymeric prodrug of BUF holds promising potential toward the treatment of somatostatin receptor 2 overexpressing breast cancer.
Collapse
Affiliation(s)
- Tao Liu
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Tingting Jia
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xia Yuan
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Cheng Liu
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jian Sun
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zhenhua Ni
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jian Xu
- Centralab, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xuhui Wang
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yi Yuan
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
36
|
Huo M, Liu Y, Wang L, Yin T, Qin C, Xiao Y, Yin L, Liu J, Zhou J. Redox-Sensitive Micelles Based on O,N-Hydroxyethyl Chitosan-Octylamine Conjugates for Triggered Intracellular Delivery of Paclitaxel. Mol Pharm 2016; 13:1750-62. [PMID: 27100204 DOI: 10.1021/acs.molpharmaceut.5b00696] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A redox-sensitive micellar system constructed from an O,N-hydroxyethyl chitosan-octylamine (HECS-ss-OA) conjugate with disulfide linkages between the hydrophobic alkyl chains and hydrophilic chitosan backbone was synthesized for triggered intracellular delivery of hydrophobic paclitaxel (PTX). In aqueous environments, conjugates formed micelles with high PTX loading (>30%). Mechanistically, the sensitivity of HECS-ss-OA micelles to reducing environments was investigated using the parameters of in vitro release and particle size. Intracellular release of nile red fluorescence alongside cytotoxicity studies further confirmed the potency of redox-sensitive micelles for intracellular drug delivery compared with redox-insensitive micelles. Additionally, an in vivo study confirmed the efficacy of PTX-loaded micelles in tumor-bearing mice with superior antitumor efficacy and diminished systemic toxicity when compared with the redox-insensitive micelles and a PTX solution. These results demonstrate the potential of redox-sensitive HECS-ss-OA micelles for intracellular trafficking of lipophilic anticancer drugs.
Collapse
Affiliation(s)
- Meirong Huo
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Yao Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Lei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Tingjie Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Chen Qin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Yanyu Xiao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Lifang Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| | - Jiyong Liu
- Department of Pharmacy, Changhai Hospital, The Second Military Medical University , Shanghai 200433, China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , 24 Tongjiaxiang, Nanjing 210009, China
| |
Collapse
|
37
|
Chen G, Jaskula-Sztul R, Harrison A, Dammalapati A, Xu W, Cheng Y, Chen H, Gong S. KE108-conjugated unimolecular micelles loaded with a novel HDAC inhibitor thailandepsin-A for targeted neuroendocrine cancer therapy. Biomaterials 2016; 97:22-33. [PMID: 27156249 DOI: 10.1016/j.biomaterials.2016.04.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/06/2016] [Accepted: 04/20/2016] [Indexed: 12/18/2022]
Abstract
Neuroendocrine (NE) cancers can cause significant patient morbidity. Besides surgery, there are no curative treatments for NE cancers and their metastases, emphasizing the need for the development of other forms of therapy. In this study, multifunctional unimolecular micelles were developed for targeted NE cancer therapy. The unimolecular micelles were formed by multi-arm star amphiphilic block copolymer poly(amidoamine)-poly(valerolactone)-poly(ethylene glycol) conjugated with KE108 peptide and Cy5 dye (abbreviated as PAMAM-PVL-PEG-KE108/Cy5). The unimolecular micelles with a spherical core-shell structure exhibited a uniform size distribution and excellent stability. The hydrophobic drug thailandepsin-A (TDP-A), a recently discovered HDAC inhibitor, was physically encapsulated into the hydrophobic core of the micelles. KE108 peptide, a somatostatin analog possessing high affinity for all five subtypes of somatostatin receptors (SSTR 1-5), commonly overexpressed in NE cancer cells, was used for the first time as an NE cancer targeting ligand. KE108 exhibited superior targeting abilities compared to other common somatostatin analogs, such as octreotide, in NE cancer cell lines. The in vitro assays demonstrated that the TDP-A-loaded, KE108-targeted micelles exhibited the best capabilities in suppressing NE cancer cell growth. Moreover, the in vivo near-infrared fluorescence imaging on NE-tumor-bearing nude mice showed that KE108-conjugated micelles exhibited the greatest tumor accumulation due to their passive targeting and active targeting capabilities. Finally, TDP-A-loaded and KE108-conjugated micelles possessed the best anticancer efficacy without detectable systemic toxicity. Thus, these novel TDP-A-loaded and KE108-conjugated unimolecular micelles offer a promising approach for targeted NE cancer therapy.
Collapse
Affiliation(s)
- Guojun Chen
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Renata Jaskula-Sztul
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - April Harrison
- Department of Surgery, University of Wisconsin-Madison, WI 53705, USA
| | | | - Wenjin Xu
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yiqiang Cheng
- University of Texas Health Sciences Center San Anto-Division, San Antonio, TX 76107, USA
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Shaoqin Gong
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
38
|
Rongbin H, Lei X, Ying L, Xiangping D, Xuan C, Lanfang L, Cuiyun Y, Yanming C, Guotao T. Synthesis and in vitro evaluation of pH-sensitive PEG-I-dC16 block polymer micelles for anticancer drug delivery. J Pharm Pharmacol 2016; 68:751-61. [DOI: 10.1111/jphp.12545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 02/14/2016] [Indexed: 11/30/2022]
Abstract
Abstract
Objectives
To develop an acid trigger release of antitumour drug delivery carriers, pH-sensitive amphiphilic poly (ethyleneglycol)-imine-benzoic-dipalmitate (PEG-I-dC16) polymers were designed and synthesized and the drug-loaded micelles were evaluated in vitro.
Methods
PEG-I-dC16 synthesized by Schiff base synthetic method and characterized by 1H-NMR. To determine the drug-loading capacity, doxorubicin (DOX) was encapsulated in the micelles using membrane dialysis method. Zeta potential, particle size, drug-loading capacity, in vitro drug release in different pH conditions and cytotoxicity evaluation of micelles were carried out comparing with non-acid liable PEG–amide–benzoic–dipalmitate (PEG-A-dC16) polymers micelles. The cellular uptake and intracellular distribution of DOX were detected by flow cytometry and confocal laser scanning microscope.
Key findings
Drug-loading capacity and encapsulation efficiency of micelle (PEG molecular weight 2k) were 12.7 ± 1.1% and 49.8 ± 2.2%, respectively. The average particle size was 72.3 ± 2.5 nm. The DOX release rate of PEG-I-dC16 micelles is much higher at pH 6.5 than at pH 7.4. DOX cellular uptake and nuclear accumulation of PEG-I-dC16 micelles were more efficiency than that of PEG-A-dC16 micelles.
Conclusion
The pH-sensitive PEG-I-dC16 micelles could be a promising drug delivery system for anticancer drugs.
Collapse
Affiliation(s)
- Huang Rongbin
- The First People's Hospital of Xiangtan City, Xiangtan, China
| | - Xiang Lei
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics University of South China, Hengyang, China
| | - Liu Ying
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics University of South China, Hengyang, China
| | - Deng Xiangping
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics University of South China, Hengyang, China
| | - Cao Xuan
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics University of South China, Hengyang, China
| | - Li Lanfang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics University of South China, Hengyang, China
| | - Yu Cuiyun
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics University of South China, Hengyang, China
| | - Chen Yanming
- Mu Dan Jiang You Bo Pharmacertical Co.Ltd, Mudanjiang, China
| | - Tang Guotao
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics University of South China, Hengyang, China
| |
Collapse
|
39
|
|
40
|
Jaskula-Sztul R, Xu W, Chen G, Harrison A, Dammalapati A, Nair R, Cheng Y, Gong S, Chen H. Thailandepsin A-loaded and octreotide-functionalized unimolecular micelles for targeted neuroendocrine cancer therapy. Biomaterials 2016; 91:1-10. [PMID: 26994874 DOI: 10.1016/j.biomaterials.2016.03.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 02/26/2016] [Accepted: 03/06/2016] [Indexed: 01/12/2023]
Abstract
Due to the overexpression of somatostatin receptors in neuroendocrine (NE) cancers, drug nanocarriers conjugated with somatostatin analogs, such as octreotide (OCT), for targeted NE cancer therapy may offer increased therapeutic efficacies and decreased adverse effects. In this study, OCT-functionalized unimolecular micelles were prepared using individual hyperbranched polymer molecules consisting of a hyperbranched polymer core (Boltorn(®) H40) and approximately 25 amphiphilic polylactide-poly(ethlyene glycol) (PLA-PEG) block copolymer arms (H40-PLA-PEG-OCH3/OCT). The resulting micelles, exhibiting a uniform core-shell shape and an average hydrodynamic diameter size of 66 nm, were loaded with thailandepsin-A (TDP-A), a relatively new naturally produced histone deacetylase (HDAC) inhibitor. In vitro studies using flow cytometry and confocal laser scanning microscopy (CLSM) demonstrated that OCT conjugation enhanced the cellular uptake of the unimolecular micelles. Consequently, TDP-A-loaded and OCT-conjugated micelles exhibited the highest cytotoxicity and caused the highest reduction of NE tumor markers. Finally, the in vivo studies on NE cancer bearing nude mice demonstrated that TDP-A-loaded and OCT-conjugated micelles possessed superior anticancer activity in comparison with other TDP-A formulations or drug alone, while showing no detectable systemic toxicity. Thus, these TDP-A-loaded and OCT-conjugated micelles offer a promising approach for targeted NE cancer therapy.
Collapse
Affiliation(s)
| | - Wenjin Xu
- Department of Biomedical Engineering and Wisconsin Institutes for Discovery, University of Wisconsin-Madison, 1550 Engineering Drive, 3144 Engineering Centers Building, Madison, WI, 53715, USA
| | - Guojun Chen
- Department of Biomedical Engineering and Wisconsin Institutes for Discovery, University of Wisconsin-Madison, 1550 Engineering Drive, 3144 Engineering Centers Building, Madison, WI, 53715, USA; Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - April Harrison
- Department of Surgery, University of Wisconsin, Madison, WI, 53705, USA
| | | | - Renu Nair
- Department of Surgery, University of Wisconsin, Madison, WI, 53705, USA
| | - Yiqiang Cheng
- University of Texas Health Sciences Center San Anto-Division, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Shaoqin Gong
- Department of Biomedical Engineering and Wisconsin Institutes for Discovery, University of Wisconsin-Madison, 1550 Engineering Drive, 3144 Engineering Centers Building, Madison, WI, 53715, USA; Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| | - Herbert Chen
- Department of Surgery, University of Alabama - Birmingham, Birmingham, 1808 7th Avenue South / Suite 502, 35233, AL, USA.
| |
Collapse
|
41
|
Zhang HY, Xu WQ, Wang YW, Omari-Siaw E, Wang Y, Zheng YY, Cao X, Tong SS, Yu JN, Xu XM. Tumor targeted delivery of octreotide-periplogenin conjugate: Synthesis, in vitro and in vivo evaluation. Int J Pharm 2016; 502:98-106. [PMID: 26899980 DOI: 10.1016/j.ijpharm.2016.02.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 02/01/2016] [Accepted: 02/14/2016] [Indexed: 12/17/2022]
Abstract
Periplogenin (PPG), a cardiac glycoside prepared from Cortex periplocae, with similar structure to bufalin, has been found to induce apoptosis in many tumor cells. However, lots of cardiac glycosides possessing strong antitumor activity in vitro have still not passed phase I clinical trials, mostly due to poor tumor selectivity and systemic toxicity. To overcome this drawback, we designed octreotide-periplogenin (OCT-PPG) conjugate by coupling PPG-succinate to the amino-terminal end of octreotide. In comparison with free PPG, the conjugate exhibited significantly stronger cytotoxicity on HepG2 cells (SSTRs overexpression) but much less toxicity in L-02 cells. After intravenous injection of OCT-PPG conjugate into H22 tumor-bearing mice, its total accumulation in tumor was 2.3 fold higher than that of free PPG, but was 0.71- and 0.84-fold lower in heart and liver, respectively, suggesting somatostatin-mediated target delivery of PPG into the tumor tissue and reduced distribution in heart and liver. In vivo studies using H22 tumor model in mice confirmed the remarkable therapeutic effect of this conjugate. These results suggested that OCT-PPG conjugate could provide a new approach for clinical application of cardiac glycosides and as a targeting agent for cancer therapy.
Collapse
Affiliation(s)
- Hui-Yun Zhang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Wen-Qian Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Yuan-Wen Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Emmanuel Omari-Siaw
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Yan Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Yuan-Yuan Zheng
- School of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Shan-Shan Tong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China
| | - Jiang-Nan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China; School of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China.
| | - Xi-Ming Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, People's Republic of China.
| |
Collapse
|
42
|
Drug delivery system targeting advanced hepatocellular carcinoma: Current and future. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:853-869. [PMID: 26772424 DOI: 10.1016/j.nano.2015.12.381] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 12/16/2015] [Accepted: 12/22/2015] [Indexed: 12/21/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) has a fairly high morbidity and is notoriously difficult to treat due to long latent period before detection, multidrug resistance and severe drug-related adverse effects from chemotherapy. Targeted drug delivery systems (DDS) that can selectively deliver therapeutic drugs into tumor sites have demonstrated a great potential in cancer treatment, which could be utilized to resolve the limitations of conventional chemotherapy. Numerous preclinical studies of DDS have been published, but targeted DDS for HCC has yet to be made for practical clinical use. Since rational targeted DDS design should take cancer-specific properties into consideration, we have reviewed the biological and physicochemical properties of HCC extensively to provide a comprehensive understanding on HCC, and recent DDS studies on HCC, aiming to find some potential targeted DDSs for HCC treatment and a meaningful platform for further development of HCC treatments. FROM THE CLINICAL EDITOR Hepatocellular carcinoma has a high incidence worldwide and is known to be multidrug resistant. Thus, intensive research is being carried out to find better chemotherapeutic agents as well as new drug delivery systems. In this article, the authors reviewed in depth the current challenges facing new drug designs and also outlined novel targeted drug delivery systems (DDS) in the fight against HCC.
Collapse
|
43
|
Woraphatphadung T, Sajomsang W, Gonil P, Treetong A, Akkaramongkolporn P, Ngawhirunpat T, Opanasopit P. pH-Responsive polymeric micelles based on amphiphilic chitosan derivatives: Effect of hydrophobic cores on oral meloxicam delivery. Int J Pharm 2016; 497:150-60. [DOI: 10.1016/j.ijpharm.2015.12.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/25/2015] [Accepted: 12/03/2015] [Indexed: 10/22/2022]
|
44
|
Lelle M, Kaloyanova S, Freidel C, Theodoropoulou M, Musheev M, Niehrs C, Stalla G, Peneva K. Octreotide-Mediated Tumor-Targeted Drug Delivery via a Cleavable Doxorubicin-Peptide Conjugate. Mol Pharm 2015; 12:4290-300. [PMID: 26524088 DOI: 10.1021/acs.molpharmaceut.5b00487] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Although recent methods for targeted drug delivery have addressed many of the existing problems of cancer therapy associated with undesirable side effects, significant challenges remain that have to be met before they find significant clinical relevance. One such area is the delicate chemical bond that is applied to connect a cytotoxic drug with targeting moieties like antibodies or peptides. Here we describe a novel platform that can be utilized for the preparation of drug-carrier conjugates in a site-specific manner, which provides excellent versatility and enables triggered release inside cancer cells. Its key feature is a cleavable doxorubicin-octreotide bioconjugate that targets overexpressed somatostatin receptors on tumor cells, where the coupling between the two components was achieved through the first cleavable disulfide-intercalating linker. The tumor targeting ability and suppression of adrenocorticotropic hormone secretion in AtT-20 cells by both octreotide and the doxorubicin hybrid were determined via a specific radioimmunoassay. Both substances reduced the hormone secretion to a similar extent, which demonstrated that the tumor homing peptide is able to interact with the relevant cell surface receptors after the attachment of the drug. Effective drug release was quickly accomplished in the presence of the physiological reducing agent glutathione. We also demonstrate the relevance of this scaffold in biological context in cytotoxicity assays with pituitary, pancreatic, and breast cancer cell lines.
Collapse
Affiliation(s)
- Marco Lelle
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Stefka Kaloyanova
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Christoph Freidel
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | | | - Michael Musheev
- Institute of Molecular Biology, Ackermannweg 4, 55128 Mainz, Germany
| | - Christof Niehrs
- Institute of Molecular Biology, Ackermannweg 4, 55128 Mainz, Germany.,Division of Molecular Embryology, DKFZ-ZMBH Alliance , 69120 Heidelberg, Germany
| | - Günter Stalla
- Max Planck Institute for Psychiatry, Kraepelinstraße 2-10, 80804 Munich, Germany
| | - Kalina Peneva
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.,Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich Schiller University Jena , Lessingstrasse 8, 07743 Jena, Germany
| |
Collapse
|
45
|
Beck P, Cui H, Hegemann JD, Marahiel MA, Krüger A, Groll M. Targeted Delivery of Proteasome Inhibitors to Somatostatin-Receptor-Expressing Cancer Cells by Octreotide Conjugation. ChemMedChem 2015; 10:1969-73. [PMID: 26471124 DOI: 10.1002/cmdc.201500449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Indexed: 12/24/2022]
Abstract
Clinical application of proteasome inhibitors (PIs) is so far limited to peripheral blood cancers due to the pronounced cytotoxicity towards all cell types. Targeted delivery of PIs could permit the treatment of other cancers along with decreasing side effects. Herein we describe the first small-molecule proteasome inhibitor conjugate for targeted delivery, created by fusing PIs to a synthetic ligand of somatostatin receptors, which are highly expressed in a variety of tumors. X-ray crystallographic studies and in vitro IC50 measurements demonstrated that addition of the cyclopeptide octreotide as a targeting vehicle does not affect the PI's binding mode. The cytotoxicity of the conjugate against somatostatin-receptor-expressing cells was up to 11-fold higher than that of a non-targeting surrogate. We have therefore established PIs as a new payload for drug conjugates and have shown that targeted delivery thereof could be a promising approach for the broader application of this FDA-approved class of compounds.
Collapse
Affiliation(s)
- Philipp Beck
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, 85748, Garching, Germany.
| | - Haissi Cui
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, 85748, Garching, Germany
| | - Julian D Hegemann
- Department of Chemistry/Biochemistry, Philipps-Universität Marburg, Hans-Meerwein-Straße 4, 35032, Marburg, Germany
| | - Mohammed A Marahiel
- Department of Chemistry/Biochemistry, Philipps-Universität Marburg, Hans-Meerwein-Straße 4, 35032, Marburg, Germany
| | - Achim Krüger
- Institute of Molecular Immunology and Experimental Oncology, Klinikum Rechts der Isar der Technische Universität München, Ismaninger Straße 22, 81675, München, Germany
| | - Michael Groll
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, 85748, Garching, Germany.
| |
Collapse
|
46
|
Nogueira E, Gomes AC, Preto A, Cavaco-Paulo A. Design of liposomal formulations for cell targeting. Colloids Surf B Biointerfaces 2015; 136:514-26. [PMID: 26454541 DOI: 10.1016/j.colsurfb.2015.09.034] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/16/2015] [Accepted: 09/18/2015] [Indexed: 01/04/2023]
Abstract
Liposomes have gained extensive attention as carriers for a wide range of drugs due to being both nontoxic and biodegradable as they are composed of substances naturally occurring in biological membranes. Active targeting for cells has explored specific modification of the liposome surface by functionalizing it with specific targeting ligands in order to increase accumulation and intracellular uptake into target cells. None of the Food and Drug Administration-licensed liposomes or lipid nanoparticles are coated with ligands or target moieties to delivery for homing drugs to target tissues, cells or subcellular organelles. Targeted therapies (with or without controlled drug release) are an emerging and relevant research area. Despite of the numerous liposomes reviews published in the last decades, this area is in constant development. Updates urgently needed to integrate new advances in targeted liposomes research. This review highlights the evolution of liposomes from passive to active targeting and challenges in the development of targeted liposomes for specific therapies.
Collapse
Affiliation(s)
- Eugénia Nogueira
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; CEB-Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Andreia C Gomes
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Ana Preto
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Artur Cavaco-Paulo
- CEB-Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal.
| |
Collapse
|
47
|
Huo M, Zhu Q, Wu Q, Yin T, Wang L, Yin L, Zhou J. Somatostatin Receptor–Mediated Specific Delivery of Paclitaxel Prodrugs for Efficient Cancer Therapy. J Pharm Sci 2015; 104:2018-2028. [DOI: 10.1002/jps.24438] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/01/2015] [Accepted: 03/05/2015] [Indexed: 12/31/2022]
|
48
|
Liang D, Wang AT, Yang ZZ, Liu YJ, Qi XR. Enhance Cancer Cell Recognition and Overcome Drug Resistance Using Hyaluronic Acid and α-Tocopheryl Succinate Based Multifunctional Nanoparticles. Mol Pharm 2015; 12:2189-202. [PMID: 25945733 DOI: 10.1021/acs.molpharmaceut.5b00129] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Multidrug resistance (MDR) presents a clinical obstacle to cancer chemotherapy. The main purpose of this study was to evaluate the potential of a hyaluronic acid (HA) and α-tocopheryl succinate (α-TOS) based nanoparticle to enhance cancer cell recognition and overcome MDR, and to explore the underlying mechanisms. A multifunctional nanoparticle, HTTP-50 NP, consisted of HA-α-TOS (HT) conjugate and d-α-tocopheryl polyethylene glycol succinate (TPGS) with docetaxel loaded in its hydrophobic core. The promoted tumor cell recognition and accumulation, cytotoxicity, and mitochondria-specific apoptotic pathways for the HTTP-50 NP were confirmed in MCF-7/Adr cells (P-gp-overexpressing cancer model), indicating that the formulated DTX and the conjugated α-TOS in the HTTP-50 NP could synergistically circumvent the acquired and intrinsic MDR in MCF-7/Adr cells. In vivo investigation on the MCF-7/Adr xenografted nude mice models confirmed that HTTP-50 NP possessed much higher tumor tissue accumulation and exhibited pronouncedly enhanced antiresistance tumor efficacy with reduced systemic toxicity compared with HTTP-0 NP and Taxotere. The mechanisms of the multifunctional HTTP-50 NP to overcome MDR and enhance antiresistance efficacy may be contributed by CD44 receptor-targeted delivery and P-gp efflux inhibition, and meanwhile to maximize antitumor efficacy by synergism of DTX and mitocan of α-TOS killing tumor cells.
Collapse
Affiliation(s)
- Desheng Liang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, People's Republic of China
| | - Ai-Ting Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, People's Republic of China
| | - Zhen-Zhen Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, People's Republic of China
| | - Yu-Jie Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, People's Republic of China
| | - Xian-Rong Qi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, People's Republic of China
| |
Collapse
|
49
|
Zhang T, Xiong H, Dahmani FZ, Sun L, Li Y, Yao L, Zhou J, Yao J. Combination chemotherapy of doxorubicin, all-trans retinoic acid and low molecular weight heparin based on self-assembled multi-functional polymeric nanoparticles. NANOTECHNOLOGY 2015; 26:145101. [PMID: 25771790 DOI: 10.1088/0957-4484/26/14/145101] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Based on the complementary effects of doxorubicin (DOX), all-trans retinoic acid (ATRA) and low molecular weight heparin (LMWH), the combination therapy of DOX, ATRA and LMWH was expected to exert the enhanced anti-tumor effects and reduce the side effects. In this study, amphiphilic LMWH-ATRA conjugate was synthesized for encapsulating the DOX. In this way, DOX, ATRA and LMWH were assembled into a single nano-system by both chemical and physical modes to obtain a novel anti-tumor targeting drug delivery system that can realize the simultaneous delivery of multiple drugs with different properties to the tumor. LMWH-ATRA nanoparticles exhibited good loading capacities for DOX with excellent physico-chemical properties, good biocompatibility, and good differentiation-inducing activity and antiangiogenic activity. The drug-loading capacity was up to 18.7% with an entrapment efficiency of 78.8%. It was also found that DOX-loaded LMWH-ATRA nanoparticles (DHR nanoparticles) could be efficiently taken up by tumor cells via endocytic pathway, and mainly distributed in cytoplasm at first, then transferred into cell nucleus. Cell viability assays suggested that DHR nanoparticles maintained the cytotoxicity effect of DOX on MCF-7 cells. Moreover, the in vivo imaging analysis indicated that DiR-loaded LMWH-ATRA nanoparticles could target the tumor more effectively as compared to free DiR. Furthermore, DHR nanoparticles possessed much higher anticancer activity and reduced side effects compared to free drugs solution. These results suggested that DHR nanoparticles could be considered as a promising targeted delivery system for combination cancer chemotherapy with lower adverse effects.
Collapse
Affiliation(s)
- Ting Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Srinivasarao M, Galliford CV, Low PS. Principles in the design of ligand-targeted cancer therapeutics and imaging agents. Nat Rev Drug Discov 2015; 14:203-19. [DOI: 10.1038/nrd4519] [Citation(s) in RCA: 476] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|