1
|
Li J, Cao Y, Zhang X, An M, Liu Y. The Application of Nano-drug Delivery System With Sequential Drug Release Strategies in Cancer Therapy. Am J Clin Oncol 2023; 46:459-473. [PMID: 37533151 DOI: 10.1097/coc.0000000000001030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Currently, multidrug combinations are often used clinically to improve the efficacy of oncology chemotherapy, but multidrug combinations often lead to multidrug resistance and decreased performance, resulting in more severe side effects than monotherapy. Therefore, sequential drug release strategies in time and space as well as nano-carriers that respond to the tumor microenvironment have been developed. First, the advantage of the sequential release strategy is that they can load multiple drugs simultaneously to meet their spatiotemporal requirements and stability, thus exerting synergistic effects of two or more drugs. Second, in some cases, sequential drug delivery of different molecular targets can improve the sensitivity of cancer cells to drugs. Control the metabolism of cancer cells, and remodel tumor vasculature. Finally, some drug combinations with built-in release control are used for sequential administration. This paper focuses on the use of nanotechnology and built-in control device to construct drug delivery carriers with different stimulation responses, thus achieving the sequential release of drugs. Therefore, the nano-sequential delivery carrier provides a new idea and platform for the therapeutic effect of various drugs and the synergistic effect among drugs.
Collapse
Affiliation(s)
- Juan Li
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | | | | | | | | |
Collapse
|
2
|
Wang C, Xu J, Zhang Y, Nie G. Emerging nanotechnological approaches to regulating tumor vasculature for cancer therapy. J Control Release 2023; 362:647-666. [PMID: 37703928 DOI: 10.1016/j.jconrel.2023.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Abnormal angiogenesis stands for one of the most striking manifestations of malignant tumor. The pathologically and structurally abnormal tumor vasculature facilitates a hostile tumor microenvironment, providing an ideal refuge exclusively for cancer cells. The emergence of vascular regulation drugs has introduced a distinctive class of therapeutics capable of influencing nutrition supply and drug delivery efficacy without the need to penetrate a series of physical barriers to reach tumor cells. Nanomedicines have been further developed for more precise regulation of tumor vasculature with the capacity of co-delivering multiple active pharmaceutical ingredients, which overall reduces the systemic toxicity and boosts the therapeutic efficacy of free drugs. Additionally, precise structure design enables the integration of specific functional motifs, such as surface-targeting ligands, droppable shells, degradable framework, or stimuli-responsive components into nanomedicines, which can improve tissue-specific accumulation, enhance tissue penetration, and realize the controlled and stimulus-triggered release of the loaded cargo. This review describes the morphological and functional characteristics of tumor blood vessels and summarizes the pivotal molecular targets commonly used in nanomedicine design, and then highlights the recent cutting-edge advancements utilizing nanotechnologies for precise regulation of tumor vasculature. Finally, the challenges and future directions of this field are discussed.
Collapse
Affiliation(s)
- Chunling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yinlong Zhang
- Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; School of Nanoscience and Engineering, School of Chemical Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; GBA National Institute for Nanotechnology Innovation, Guangzhou 510530, China.
| |
Collapse
|
3
|
Xu X, Liu A, Liu S, Ma Y, Zhang X, Zhang M, Zhao J, Sun S, Sun X. Application of molecular dynamics simulation in self-assembled cancer nanomedicine. Biomater Res 2023; 27:39. [PMID: 37143168 PMCID: PMC10161522 DOI: 10.1186/s40824-023-00386-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Self-assembled nanomedicine holds great potential in cancer theragnostic. The structures and dynamics of nanomedicine can be affected by a variety of non-covalent interactions, so it is essential to ensure the self-assembly process at atomic level. Molecular dynamics (MD) simulation is a key technology to link microcosm and macroscale. Along with the rapid development of computational power and simulation methods, scientists could simulate the specific process of intermolecular interactions. Thus, some experimental observations could be explained at microscopic level and the nanomedicine synthesis process would have traces to follow. This review not only outlines the concept, basic principle, and the parameter setting of MD simulation, but also highlights the recent progress in MD simulation for self-assembled cancer nanomedicine. In addition, the physicochemical parameters of self-assembly structure and interaction between various assembled molecules under MD simulation are also discussed. Therefore, this review will help advanced and novice researchers to quickly zoom in on fundamental information and gather some thought-provoking ideas to advance this subfield of self-assembled cancer nanomedicine.
Collapse
Affiliation(s)
- Xueli Xu
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Ao Liu
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Shuangqing Liu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yanling Ma
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Xinyu Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Meng Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Jinhua Zhao
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Shuo Sun
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, 02115, USA
| | - Xiao Sun
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| |
Collapse
|
4
|
Yuan B, Liu Y, Lv M, Sui Y, Hou S, Yang T, Belhadj Z, Zhou Y, Chang N, Ren Y, Sun C. Virus-like particle-based nanocarriers as an emerging platform for drug delivery. J Drug Target 2023; 31:433-455. [PMID: 36940208 DOI: 10.1080/1061186x.2023.2193358] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
New nanocarrier technologies are emerging, and they have great potential for improving drug delivery, targeting efficiency, and bioavailability. Virus-like particles (VLPs) are natural nanoparticles from animal and plant viruses and bacteriophages. Hence, VLPs present several great advantages, such as morphological uniformity, biocompatibility, reduced toxicity, and easy functionalisation. VLPs can deliver many active ingredients to the target tissue and have great potential as a nanocarrier to overcome the limitations associated with other nanoparticles. This review will focus primarily on the construction and applications of VLPs, particularly as a novel nanocarrier to deliver active ingredients. Herein, the main methods for the construction, purification, and characterisation of VLPs, as well as various VLP-based materials used in delivery systems are summarised. The biological distribution of VLPs in drug delivery, phagocyte-mediated clearance, and toxicity are also discussed.
Collapse
Affiliation(s)
| | - Yang Liu
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| | - Meilin Lv
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Yilei Sui
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Shenghua Hou
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Tinghui Yang
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Zakia Belhadj
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yulong Zhou
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Naidan Chang
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Yachao Ren
- Harbin Medical University-Daqing, Daqing 163319, China.,School of Chemistry and Chemical Engineering, Tianjin University of Technology, tianjin, 300000, China
| | | |
Collapse
|
5
|
Xie L, Liu R, Chen X, He M, Zhang Y, Chen S. Micelles Based on Lysine, Histidine, or Arginine: Designing Structures for Enhanced Drug Delivery. Front Bioeng Biotechnol 2021; 9:744657. [PMID: 34646819 PMCID: PMC8503256 DOI: 10.3389/fbioe.2021.744657] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/30/2021] [Indexed: 01/10/2023] Open
Abstract
Natural amino acids and their derivatives are excellent building blocks of polymers for various biomedical applications owing to the non-toxicity, biocompatibility, and ease of multifunctionalization. In the present review, we summarized the common approaches to designing and constructing functional polymeric micelles based on basic amino acids including lysine, histidine, and arginine and highlighted their applications as drug carriers for cancer therapy. Different polypeptide architectures including linear polypeptides and dendrimers were developed for efficient drug loading and delivery. Besides, polylysine- and polyhistidine-based micelles could enable pH-responsive drug release, and polyarginine can realize enhanced membrane penetration and gas therapy by generating metabolites of nitric oxide (NO). It is worth mentioning that according to the structural or functional characteristics of basic amino acids and their derivatives, key points for designing functional micelles with excellent drug delivery efficiency are importantly elaborated in order to pave the way for exploring micelles based on basic amino acids.
Collapse
Affiliation(s)
- Li Xie
- School of Medicine and Nursing, Chengdu University, Chengdu, China
| | - Rong Liu
- School of Medicine and Nursing, Chengdu University, Chengdu, China
| | - Xin Chen
- School of Medicine and Nursing, Chengdu University, Chengdu, China
| | - Mei He
- School of Medicine and Nursing, Chengdu University, Chengdu, China
| | - Yi Zhang
- School of Medicine and Nursing, Chengdu University, Chengdu, China
| | - Shuyi Chen
- School of Medicine and Nursing, Chengdu University, Chengdu, China
| |
Collapse
|
6
|
Prencipe F, Diaferia C, Rossi F, Ronga L, Tesauro D. Forward Precision Medicine: Micelles for Active Targeting Driven by Peptides. Molecules 2021; 26:4049. [PMID: 34279392 PMCID: PMC8271712 DOI: 10.3390/molecules26134049] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Precision medicine is based on innovative administration methods of active principles. Drug delivery on tissue of interest allows improving the therapeutic index and reducing the side effects. Active targeting by means of drug-encapsulated micelles decorated with targeting bioactive moieties represents a new frontier. Between the bioactive moieties, peptides, for their versatility, easy synthesis and immunogenicity, can be selected to direct a drug toward a considerable number of molecular targets overexpressed on both cancer vasculature and cancer cells. Moreover, short peptide sequences can facilitate cellular intake. This review focuses on micelles achieved by self-assembling or mixing peptide-grafted surfactants or peptide-decorated amphiphilic copolymers. Nanovectors loaded with hydrophobic or hydrophilic cytotoxic drugs or with gene silence sequences and externally functionalized with natural or synthetic peptides are described based on their formulation and in vitro and in vivo behaviors.
Collapse
Affiliation(s)
- Filippo Prencipe
- Institute of Crystallography (IC) CNR, Via Amendola 122/o, 70126 Bari, Italy
| | - Carlo Diaferia
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", via Mezzocannone 16, 80134 Naples, Italy
| | - Filomena Rossi
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", via Mezzocannone 16, 80134 Naples, Italy
| | - Luisa Ronga
- Institut des Sciences Analytiques et de Physico-Chimie Pour l'Environnement et les Matériaux, Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, 64053 Pau, France
| | - Diego Tesauro
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", via Mezzocannone 16, 80134 Naples, Italy
| |
Collapse
|
7
|
Mirzavi F, Barati M, Soleimani A, Vakili-Ghartavol R, Jaafari MR, Soukhtanloo M. A review on liposome-based therapeutic approaches against malignant melanoma. Int J Pharm 2021; 599:120413. [PMID: 33667562 DOI: 10.1016/j.ijpharm.2021.120413] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 01/14/2023]
Abstract
Melanoma is a highly aggressive form of skin cancer with a very poor prognosis and excessive resistance to current conventional treatments. Recently, the application of the liposomal delivery system in the management of skin melanoma has been widely investigated. Liposomal nanocarriers are biocompatible and less toxic to host cells, enabling the efficient and safe delivery of different therapeutic agents into the tumor site and further promoting their antitumor activities. Therefore, the liposomal delivery system effectively increases the success of current melanoma therapies and overcomes resistance. In this review, we present an overview of liposome-based targeted drug delivery methods and highlight recent advances towards the development of liposome-based carriers for therapeutic genes. We also discuss the new insights regarding the efficacy and clinical significance of combinatorial treatment of liposomal formulations with immunotherapy and conventional therapies in melanoma patients for a better understanding and successfully managing cancer.
Collapse
Affiliation(s)
- Farshad Mirzavi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Barati
- Department of Medical Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Anvar Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roghayyeh Vakili-Ghartavol
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Rajora AK, Ravishankar D, Zhang H, Rosenholm JM. Recent Advances and Impact of Chemotherapeutic and Antiangiogenic Nanoformulations for Combination Cancer Therapy. Pharmaceutics 2020; 12:pharmaceutics12060592. [PMID: 32630584 PMCID: PMC7356724 DOI: 10.3390/pharmaceutics12060592] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
Traditional chemotherapy, along with antiangiogenesis drugs (combination cancer therapy), has shown reduced tumor recurrence and improved antitumor effects, as tumor growth and metastasis are often dependent on tumor vascularization. However, the effect of combination chemotherapy, including synergism and additive and even antagonism effects, depends on drug combinations in an optimized ratio. Hence, nanoformulations are ideal, demonstrating a great potential for the combination therapy of chemo-antiangiogenesis for cancer. The rationale for designing various nanocarriers for combination therapy is derived from organic (polymer, lipid), inorganic, or hybrid materials. In particular, hybrid nanocarriers that consist of more than one material construct provide flexibility for different modes of entrapment within the same carrier—e.g., physical adsorption, encapsulation, and chemical conjugation strategies. These multifunctional nanocarriers can thus be used to co-deliver chemo- and antiangiogenesis drugs with tunable drug release at target sites. Hence, this review attempts to survey the most recent advances in nanoformulations and their impact on cancer treatment in a combined regimen—i.e., conventional cytotoxic and antiangiogenesis agents. The mechanisms and site-specific co-delivery strategies are also discussed herein, along with future prospects.
Collapse
Affiliation(s)
- Amit Kumar Rajora
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland;
- Correspondence: (A.K.R.); (J.M.R.)
| | - Divyashree Ravishankar
- Bioscience Department, Sygnature Discovery, Bio City, Pennyfoot St, Nottingham NG1 1GR, UK;
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland;
- Turku Bioscience Center, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Jessica M. Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland;
- Correspondence: (A.K.R.); (J.M.R.)
| |
Collapse
|
9
|
Tao N, Liu Y, Wu Y, Li X, Li J, Sun X, Chen S, Liu YN. Minimally Invasive Antitumor Therapy Using Biodegradable Nanocomposite Micellar Hydrogel with Functionalities of NIR-II Photothermal Ablation and Vascular Disruption. ACS APPLIED BIO MATERIALS 2020; 3:4531-4542. [DOI: 10.1021/acsabm.0c00465] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Na Tao
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P.R. China
| | - Yandi Liu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P.R. China
| | - Yingjiao Wu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P.R. China
| | - Xilong Li
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P.R. China
| | - Juan Li
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P.R. China
| | - Xiaoyi Sun
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P.R. China
| | - Shu Chen
- Key Laboratory of Theoretical Organic Chemistry and Function Molecule of Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, P.R. China
| | - You-Nian Liu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P.R. China
| |
Collapse
|
10
|
Han W, Shi L, Xie B, Wan J, Ren L, Wang Y, Chen X, Wang H. Supramolecular Engineering of Molecular Inhibitors in an Adaptive Cytotoxic Nanoparticle for Synergistic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:1707-1720. [PMID: 31816241 DOI: 10.1021/acsami.9b20178] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Combinatorial regimens that rationally pair molecular inhibitors with standard cytotoxic chemotherapeutics are used to improve therapeutic outcomes. Simultaneously engineering these therapies within a single nanocarrier that spans cytotoxic, antiangiogenic, and anti-invasive mechanisms and that enables the delivery of unique drug combinations remains a technical challenge. In this study, we developed a simple and broadly applicable strategy in which ultrastable cytotoxic nanoparticles with an established excellent antitumor efficacy and π-rich inner core structure supramolecularly stabilized the antiangiogenic molecular inhibitor apatinib to create a synergistic drug delivery system (termed sTKI-pSN38). This small-sized nanoparticle accomplished the sequential release of both encapsulated drugs to exert antimetastatic, antivascular, and cytotoxic activities simultaneously. In xenograft models of hepatocellular carcinoma, a single intravenous administration of sTKI-pSN38 elicited robust and durable tumor reduction and suppressed metastasis to lymph nodes. Interestingly, sTKI-pSN38 treatment alleviated intratumoral hypoxia, which could contribute to impaired tumor metastasis and reduced drug resistance. Collectively, this nanotherapeutic platform offers a new strategy for cancer therapy by simply engineering a drug cocktail in conventional nanoparticles and by enabling the spatiotemporal modulation of drug release to enhance the synergy of the combined drugs.
Collapse
Affiliation(s)
- Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| | - Linlin Shi
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| | - Binbin Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| | - Jianqin Wan
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , 310003 , PR China
| | - Lulu Ren
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| | - Yuchen Wang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , 310003 , PR China
| | - Xiaona Chen
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , 310003 , PR China
| | - Hangxiang Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine , Zhejiang University , Hangzhou , 310016 , PR China
| |
Collapse
|
11
|
Sumer Bolu B, Golba B, Sanyal A, Sanyal R. Trastuzumab targeted micellar delivery of docetaxel using dendron–polymer conjugates. Biomater Sci 2020; 8:2600-2610. [DOI: 10.1039/c9bm01764j] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Incorporation of a therapeutic antibody into nanosized drug delivery systems can improve their target specificity.
Collapse
Affiliation(s)
- Burcu Sumer Bolu
- Department of Chemistry
- Bogazici University
- Istanbul
- Turkey
- Center for Life Sciences and Technologies
| | - Bianka Golba
- Department of Chemistry
- Bogazici University
- Istanbul
- Turkey
| | - Amitav Sanyal
- Department of Chemistry
- Bogazici University
- Istanbul
- Turkey
- Center for Life Sciences and Technologies
| | - Rana Sanyal
- Department of Chemistry
- Bogazici University
- Istanbul
- Turkey
- Center for Life Sciences and Technologies
| |
Collapse
|
12
|
Varshosaz J, Hassanzadeh F, Hashemi-Beni B, Minaiyan M, Enteshari S. Tissue Distribution and Systemic Toxicity Evaluation of Raloxifene Targeted Polymeric Micelles of Poly (Styrene-Maleic Acid)-Poly (Amide- Ether-Ester-Imide)-Poly (Ethylene Glycol) Loaded With Docetaxel in Breast Cancer Bearing Mice. Recent Pat Anticancer Drug Discov 2019; 14:280-291. [PMID: 31538904 DOI: 10.2174/1574892814666190919163731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 01/30/2023]
Abstract
BACKGROUND Due to the low water solubility of Docetaxel (DTX), it is formulated with ethanol and Tween 80 with lots of side effects. For this reason, special attention has been paid to formulate it in new drug nano-carriers. OBJECTIVE The goal of this study was to evaluate the safety, antitumor activity and tissue distribution of the novel synthesized Raloxifene (RA) targeted polymeric micelles. METHODS DTX-loaded RA-targeted polymeric micelles composed of poly(styrene-maleic acid)- poly(amide-ether-ester-imide)-poly(ethylene glycol) (SMA-PAEE-PEG) were prepared and their antitumor activity was studied in MC4-L2 tumor-bearing mice compared with non-targeted micelles and free DTX. Safety of the micelles was studied by Hematoxylin and Eosin (H&E) staining of tumors and major organs of the mice. The drug accumulation in the tumor and major organs was measured by HPLC method. RESULTS The results showed better tumor growth inhibition and increased survival of mice treated with DTX-loaded in targeted micelles compared to the non-targeted micelles and free DTX. Histopathological studies, H&E staining of tumors and immunohistochemical examination showed the potential of DTX-loaded RA-targeted micelles to inhibit tumor cells proliferation. The higher accumulation of the DTX in the tumor tissue after injection of the micelles compared to the free DTX may indicate the higher uptake of the targeted micelles by the G-Protein-Coupled Estrogen Receptors (GPER). CONCLUSION The results indicate that RA-conjugated polymeric micelles may be a strong and effective drug delivery system for DTX therapy and uptake of the drug into tumor cells, and overcome the disadvantages and side effects of conventional DTX.
Collapse
Affiliation(s)
- Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farshid Hassanzadeh
- Department of Pharmaceutical Chemistry, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batool Hashemi-Beni
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Minaiyan
- Department of Pharmacology, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeedeh Enteshari
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
13
|
Sun M, Qian Q, Shi L, Xu L, Liu Q, Zhou L, Zhu X, Yue JM, Yan D. Amphiphilic drug-drug conjugate for cancer therapy with combination of chemotherapeutic and antiangiogenesis drugs. Sci China Chem 2019. [DOI: 10.1007/s11426-019-9602-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
14
|
Nik ME, Momtazi-Borojeni AA, Zamani P, Navashenaq JG, Iranshahi M, Jaafari MR, Malaekeh-Nikouei B. Targeted-nanoliposomal combretastatin A4 (CA-4) as an efficient antivascular candidate in the metastatic cancer treatment. J Cell Physiol 2019; 234:14721-14733. [PMID: 30697744 DOI: 10.1002/jcp.28230] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 01/03/2019] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
A number of antiangiogenic drugs have been approved by the Food and Drug Administration which are used in cancer therapy, and variety of other agents in several stages of clinical development or in preclinical assessment. Among these, combretastatin A4 (CA-4) is an under-researched inhibitor of angiogenesis that shows potential activity in the treatment of advanced tumors with migration capacity. However, its clinical application has been limited due to poor water solubility, low bioavailability, rapid metabolism, and systemic elimination. During the last decade, numerous investigations have been done to overcome these problems by using different CA-4 delivery systems or developing produgs of CA-4 or its structural analogs. Nevertheless, these strategies could not be efficient out of the undesired side effects on normal tissues. Nanoliposomal CA-4 not only benefits from the advantage of using liposomal drugs as opposed to free drugs but also can accumulate in the tumor site via specific targeting ligands, which leads to efficient targeting and enhancement of bioavailability. To the best of our knowledge, we consider an important attempt to understand different factors that might influence the CA-4 loading and release pattern of liposomes and the consequent results in tumor therapy. In this review, we shed light on various studied liposomal CA-4 formulations showing application thereof in cancer treatment.
Collapse
Affiliation(s)
- Maryam Ebrahimi Nik
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Abbas Momtazi-Borojeni
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jamshid Gholizadeh Navashenaq
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunogenetic and Cell Culture, Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bizhan Malaekeh-Nikouei
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Yang Y, Meng Y, Ye J, Xia X, Wang H, Li L, Dong W, Jin D, Liu Y. Sequential delivery of VEGF siRNA and paclitaxel for PVN destruction, anti-angiogenesis, and tumor cell apoptosis procedurally via a multi-functional polymer micelle. J Control Release 2018; 287:103-120. [PMID: 30144476 DOI: 10.1016/j.jconrel.2018.08.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/02/2018] [Accepted: 08/20/2018] [Indexed: 01/24/2023]
Abstract
Co-delivery of chemotherapy drugs and VEGF siRNA (siVEGF) to control tumor growth has been a research hotspot for improving cancer treatment. Current systems co-deliver siVEGF and chemo drugs into tumor cells simultaneously. Although effective, these systems do not flow to the abnormal blood vessels around tumor cells (vascular niche, PVN), which play an important role in the metastasis and deterioration of the tumor. Thus, we custom-synthesized triblock copolymer poly(ε-caprolactone)-polyethyleneglycol-poly(L-histidine) (PCL-PEG-PHIS) with previously synthesized folate-PEG-PHIS to construct a targeted multifunctional polymer micelle (PTX/siVEGF-CPPs/TMPM) to sequentially deliver siVEGF-CPPs (disulfide bond-linked siVEGF and cell-penetrating peptides) and paclitaxel (PTX). The sequential delivery vesicles showed the anticipated three-layered TEM structure and dual-convertible (surface charge- and particle size-reversible) features in the tumor environment (pH 6.5), which guaranteed the sequential release of siVEGF-CPPs and PTX in the tumor extracellular environment and tumor cells, respectively. To mimic the in vivo tumor environment, a double cell model was employed by co-culturing HUVECs and MCF-7 cells. Improved cell endocytosis efficiency, VEGF gene silence efficacy, and in vitro anti-proliferation activity were achieved. An in vivo study on MCF-7 tumor-bearing female nude mice also indicated that sequential delivery vesicles could lead to significant induction of tumor cell apoptosis, loss of VEGF expression, and destruction of tumor blood vessels (PVN and neovascularization). These sequential delivery vesicles show potential as an effective co-delivery platform for siVEGF and chemo drugs to improve cancer therapy efficacy.
Collapse
Affiliation(s)
- Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Yingying Meng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Xuejun Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Lin Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Wujun Dong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Dujia Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China; Beijing Key laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
16
|
Tian F, Dahmani FZ, Qiao J, Ni J, Xiong H, Liu T, Zhou J, Yao J. A targeted nanoplatform co-delivering chemotherapeutic and antiangiogenic drugs as a tool to reverse multidrug resistance in breast cancer. Acta Biomater 2018; 75:398-412. [PMID: 29874597 DOI: 10.1016/j.actbio.2018.05.050] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 05/10/2018] [Accepted: 05/30/2018] [Indexed: 12/31/2022]
Abstract
Several obstacles are currently impeding the successful treatment of breast cancer, namely impaired drug accumulation into the tumor site, toxicity to normal cells and narrow therapeutic index of chemotherapy, multidrug resistance (MDR) and the metastatic spread of cancer cells through the blood and lymphatic vessels. In this regard, we designed a novel multifunctional nano-sized drug delivery system based on LyP-1 peptide-modified low-molecular-weight heparin-quercetin conjugate (PLQ). This nanosystem was developed for targeted co-delivery of multiple anticancer drugs to p32-overexpressing tumor cells and peritumoral lymphatic vessels, using LyP-1 peptide as active targeting ligand, with the aim to achieve a targeted combinatorial chemo/angiostatic therapy and MDR reversal. The cellular uptake of PLQ nanoparticles by p32-overexpressing breast cancer cells was significantly higher than nonfunctionalized nanoparticles. Besides, the anti-angiogenic activity of PLQ nanoparticles was proven by the effective inhibition of the bFGF-induced neovascularization in subcutaneous Matrigel plugs. More importantly, PLQ/GA nanoparticles with better targeting ability toward p32-positive tumors, displayed a high antitumor outcome by inhibition of tumor cells proliferation and angiogenesis. Immunohistochemistry and western blot assay showed that PLQ/GA nanoparticles significantly disrupted the lymphatic formation of tumor, and inhibited the P-glycoprotein (P-gp) expression in MCF-7 tumor cells, respectively. In conclusion, PLQ/GA nanoparticles provide a synergistic strategy for effective targeted co-delivery of chemotherapeutic and antiangiogenic agents and reversing MDR and metastasis in breast cancer. STATEMENT OF SIGNIFICANCE Herein, we successfully developed a novel amphiphilic nanomaterial, LyP-1-LMWH-Qu (PLQ) conjugate, consisting of a tumor-targeting moiety LyP-1, a hydrophobic quercetin (a multidrug resistance [MDR]-reversing drug) inner core, and a hydrophilic low-molecular-weight heparin (an antiangiogenic agent) outer shell for encapsulating and delivering a hydrophobic chemotherapeutic agent (gambogic acid). This versatile nanoplatform with multiple targeted features, i.e., dual chemo/angiostatic effects, destruction ability of the peritumoral lymphatic vessels, and reversal of MDR, resulted in a significantly stronger antitumor efficacy and lower toxic side effect than those of nontargeted nanoparticles and the free drug solution. Therefore, this versatile nanosystem might provide a novel insight for the treatment and palliation of breast cancer by targeted co-delivery of chemo/antiangiogenic agents and reversing MDR and metastasis.
Collapse
|
17
|
Wang J, Li S, Han Y, Guan J, Chung S, Wang C, Li D. Poly(Ethylene Glycol)-Polylactide Micelles for Cancer Therapy. Front Pharmacol 2018; 9:202. [PMID: 29662450 PMCID: PMC5890116 DOI: 10.3389/fphar.2018.00202] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/22/2018] [Indexed: 12/21/2022] Open
Abstract
For the treatment of malignancy, many therapeutic agents, including small molecules, photosensitizers, immunomodulators, proteins and genes, and so forth, have been loaded into nanocarriers for controllable cancer therapy. Among these nanocarriers, polymeric micelles have been considered as one of the most promising nanocarriers, some of which have already been applied in different stages of clinical trials. The successful advantages of polymeric micelles from bench to bedside are due to their special core/shell structures, which can carry specific drugs in certain disease conditions. Particularly, poly(ethylene glycol)–polylactide (PEG–PLA) micelles have been considered as one of the most promising platforms for drug delivery. The PEG shell effectively prevents the adsorption of proteins and phagocytes, thereby evidently extending the blood circulation period. Meanwhile, the hydrophobic PLA core can effectively encapsulate many therapeutic agents. This review summarizes recent advances in PEG–PLA micelles for the treatment of malignancy. In addition, future perspectives for the development of PEG–PLA micelles as drug delivery systems are also presented.
Collapse
Affiliation(s)
- Jixue Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Shengxian Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Yuping Han
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jingjing Guan
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shirley Chung
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Chunxi Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Di Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
18
|
Ren Y, Zhan C, Gao J, Zhang M, Wei X, Ying M, Liu Z, Lu W. A d-Peptide Ligand of Integrins for Simultaneously Targeting Angiogenic Blood Vasculature and Glioma Cells. Mol Pharm 2018; 15:592-601. [DOI: 10.1021/acs.molpharmaceut.7b00944] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yachao Ren
- Department of Pharmaceutics, School of Pharmacy & Key Laboratory of Smart Drug Delivery of the Ministry of Education, Fudan University, Shanghai 201203, China
- State
Key Laboratory of Medical Neurobiology and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai 200032, China
- Harbin Medical University, Harbin 1500813, China
| | - Changyou Zhan
- Department of Pharmaceutics, School of Pharmacy & Key Laboratory of Smart Drug Delivery of the Ministry of Education, Fudan University, Shanghai 201203, China
- State
Key Laboratory of Medical Neurobiology and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai 200032, China
- Department
of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- State
Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China
| | - Jie Gao
- Department of Pharmaceutics, School of Pharmacy & Key Laboratory of Smart Drug Delivery of the Ministry of Education, Fudan University, Shanghai 201203, China
- State
Key Laboratory of Medical Neurobiology and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Mingfei Zhang
- Department of Pharmaceutics, School of Pharmacy & Key Laboratory of Smart Drug Delivery of the Ministry of Education, Fudan University, Shanghai 201203, China
- State
Key Laboratory of Medical Neurobiology and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Xiaoli Wei
- Department of Pharmaceutics, School of Pharmacy & Key Laboratory of Smart Drug Delivery of the Ministry of Education, Fudan University, Shanghai 201203, China
- State
Key Laboratory of Medical Neurobiology and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Man Ying
- Department of Pharmaceutics, School of Pharmacy & Key Laboratory of Smart Drug Delivery of the Ministry of Education, Fudan University, Shanghai 201203, China
- State
Key Laboratory of Medical Neurobiology and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Zining Liu
- Department of Pharmaceutics, School of Pharmacy & Key Laboratory of Smart Drug Delivery of the Ministry of Education, Fudan University, Shanghai 201203, China
- State
Key Laboratory of Medical Neurobiology and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy & Key Laboratory of Smart Drug Delivery of the Ministry of Education, Fudan University, Shanghai 201203, China
- State
Key Laboratory of Medical Neurobiology and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai 200032, China
- Minhang Branch,
Zhongshan Hospital, and Institute of Fudan-Minghang Academic Health
System, Minghang Hospital, Fudan University, Shanghai 201199, China
- Institutes of Integrative Medicine of Fudan University, Shanghai 200040, China
| |
Collapse
|
19
|
Bolu BS, Golba B, Boke N, Sanyal A, Sanyal R. Designing Dendron–Polymer Conjugate Based Targeted Drug Delivery Platforms with a “Mix-and-Match” Modularity. Bioconjug Chem 2017; 28:2962-2975. [DOI: 10.1021/acs.bioconjchem.7b00595] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Burcu Sumer Bolu
- Department
of Chemistry and ‡Center for Life Sciences and Technologies, Bogazici University, Istanbul, 34342, Turkey
| | - Bianka Golba
- Department
of Chemistry and ‡Center for Life Sciences and Technologies, Bogazici University, Istanbul, 34342, Turkey
| | - Nazli Boke
- Department
of Chemistry and ‡Center for Life Sciences and Technologies, Bogazici University, Istanbul, 34342, Turkey
| | - Amitav Sanyal
- Department
of Chemistry and ‡Center for Life Sciences and Technologies, Bogazici University, Istanbul, 34342, Turkey
| | - Rana Sanyal
- Department
of Chemistry and ‡Center for Life Sciences and Technologies, Bogazici University, Istanbul, 34342, Turkey
| |
Collapse
|
20
|
Thermo-sensitive polypeptide hydrogel for locally sequential delivery of two-pronged antitumor drugs. Acta Biomater 2017; 58:44-53. [PMID: 28576715 DOI: 10.1016/j.actbio.2017.05.053] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/14/2017] [Accepted: 05/30/2017] [Indexed: 11/22/2022]
Abstract
In the synergistic treatment with cytotoxic drug and vascular disrupting agent, the order of drug release shows great importance to enhance the antitumor efficacy. When vascular disrupting agent is firstly administrated, the reduced blood supply and overexpressed hypoxia-inducible factor-1α greatly limit the efficiency of chemotherapy. In this work, an injectable thermo-sensitive polypeptide hydrogel was firstly developed for the locally sequential delivery of hydrophilic doxorubicin (DOX, a cytotoxic agent) and hydrophobic combretastatin A4 (CA4, a vascular disrupting drug). The aqueous solution of polypeptide at low temperature transformed into hydrogel under the body temperature after subcutaneous injection and completely degraded after four weeks with excellent biocompatibility. DOX and CA4 were co-loaded into the hydrogel, and the release of DOX showed much faster than that of CA4 due to their difference in water solubility. The superior inhibition of tumor volume after treatment with DOX and CA4 co-loaded hydrogel occurred in the treatment of grafted mouse U14 cervical tumor compared with both free drugs and single drug-loaded hydrogels. In addition, the co-loaded hydrogel obtained enhanced apoptosis of tumor cells, significant shutdown of blood vessels, and wholly regional tumor apoptosis, which indicated the eradication of solid tumor. Moreover, treatments with the drug-loaded hydrogels showed negligible damage to normal tissues, suggesting their low systemic toxicity. The locally sequential delivery system had great potential for in situ synergistic chemotherapy. STATEMENT OF SIGNIFICANCE The release order makes great difference in the synergistic efficacies of cytotoxic drug and vascular disrupting agent. When cytotoxic drug is administrated before vascular disrupting agent, an eradication of tumor might be obtained. On the contrary, the antitumor efficiency will be greatly hindered by limited penetration of later cytotoxic drug and drug resistant induced by vascular disrupting agent. Therefore, the adjustment of the delivery behaviors of such two-pronged agents in one platform was significant for their efficiently synergistic chemotherapy. The present study originally provides a convenient strategy and an advanced sample for sequential administration of cytotoxic drug and vascular disrupting agent in one platform based on their water solubility to achieve upregulated efficacy and safety.
Collapse
|
21
|
Nanoformulation-based sequential combination cancer therapy. Adv Drug Deliv Rev 2017; 115:57-81. [PMID: 28412324 DOI: 10.1016/j.addr.2017.04.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 04/06/2017] [Accepted: 04/10/2017] [Indexed: 01/07/2023]
Abstract
Although combining two or more treatments is regarded as an indispensable approach for effectively treating cancer, the traditional cocktail-based combination therapies are seriously limited by coordination issues that fail to account for differences in the pharmacokinetics and action sites of each drug. The careful manipulation of dosing regimens, such as by the sequential application of combination treatments, may satisfy the temporal and spatial needs of each drug and achieve successful combination antitumor therapy. Nanotechnology-based carriers might be the best tools for sequential combination therapy, as they can be loaded with multiple cargos and may provide targeted and sustained delivery to target tumor cells. Single nanoformulations capable of sequentially releasing drugs have shown synergistic anticancer activity, such as by sensitizing tumor cells through cascaded drug delivery or remodeling the tumor vasculature and microenvironment to enhance the tumor distribution of nanotherapeutics. This review highlights the use of nanotechnology-based multistage drug delivery for cancer treatment, focusing on the ability of such formulations to enhance antitumor efficacy by applying sequential treatment and modulating dosing regimens, which are challenges currently being faced in the clinic.
Collapse
|
22
|
Liu T, Zhang D, Song W, Tang Z, Zhu J, Ma Z, Wang X, Chen X, Tong T. A poly(l-glutamic acid)-combretastatin A4 conjugate for solid tumor therapy: Markedly improved therapeutic efficiency through its low tissue penetration in solid tumor. Acta Biomater 2017; 53:179-189. [PMID: 28167300 DOI: 10.1016/j.actbio.2017.02.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 01/13/2023]
Abstract
Combretastatin A4 (CA4) is a leading agent in vascular disrupting strategies for tumor therapy. Although many small-molecule prodrugs of CA4 have been developed to improve its solubility, the overall therapeutic efficiency is moderate. A key reason for this is the reversible effect that CA4 has on tubulin as well as its rapid clearance from plasma and tissues. In this study, we proposed a poly(l-glutamic acid)-CA4 conjugate (PLG-CA4) nanomedicine to fulfill the requirements for fully liberating the potential of CA4 on tumor therapy. Enhanced accumulation and retention of CA4 in tumor tissue, especially, high distribution and gradual release around tumor blood vessels resulted in prolonged vascular disruption and markedly enhanced therapeutic efficiency. We examined and compared the therapeutic effect of PLG-CA4 and commercial combretastatin-A4 phosphate (CA4P) in a murine colon C26 tumor. PLG-CA4 showed significantly prolonged retention in plasma and tumor tissue. Most importantly, the PLG-CA4 was mainly distributed around the tumor vessels because of its low tissue penetration in solid tumor. Pathology tests showed that PLG-CA4 treatment resulted in persistent vascular disruption and tumor damage 72h after a single injection, this in contrast to CA4P treatment, which showed quick relapse at an equal dose. Tumor suppression tests showed that PLG-CA4 treatment resulted in a tumor suppression rate of 74%, which indicates a significant advantage when compared to tumor suppression rate of the CA4P group, which was 24%. This is the first time that an advantage of the polymeric CA4 nanomedicine with low tissue penetration for solid tumor therapy has been shown. Thus, the results presented in this study provide a new idea for enhancing the tumor therapeutic effect of vascular disrupting agents. STATEMENT OF SIGNIFICANCE Nanomedicine usually has low tissue penetration in solid tumors, which limits the efficacy of nanomedicine in most cases. But herein, we demonstrate a nanosized vascular disruptive agent (VDA) PLG-CA4 has supper advantages over small molecular combretastatin-A4 phosphate (CA4P) because the PLG-CA4 was mainly distributed around the tumor vessels due to its low tissue penetration in solid tumor.
Collapse
|
23
|
Pelaz B, Alexiou C, Alvarez-Puebla RA, Alves F, Andrews AM, Ashraf S, Balogh LP, Ballerini L, Bestetti A, Brendel C, Bosi S, Carril M, Chan WCW, Chen C, Chen X, Chen X, Cheng Z, Cui D, Du J, Dullin C, Escudero A, Feliu N, Gao M, George M, Gogotsi Y, Grünweller A, Gu Z, Halas NJ, Hampp N, Hartmann RK, Hersam MC, Hunziker P, Jian J, Jiang X, Jungebluth P, Kadhiresan P, Kataoka K, Khademhosseini A, Kopeček J, Kotov NA, Krug HF, Lee DS, Lehr CM, Leong KW, Liang XJ, Ling Lim M, Liz-Marzán LM, Ma X, Macchiarini P, Meng H, Möhwald H, Mulvaney P, Nel AE, Nie S, Nordlander P, Okano T, Oliveira J, Park TH, Penner RM, Prato M, Puntes V, Rotello VM, Samarakoon A, Schaak RE, Shen Y, Sjöqvist S, Skirtach AG, Soliman MG, Stevens MM, Sung HW, Tang BZ, Tietze R, Udugama BN, VanEpps JS, Weil T, Weiss PS, Willner I, Wu Y, Yang L, Yue Z, Zhang Q, Zhang Q, Zhang XE, Zhao Y, Zhou X, Parak WJ. Diverse Applications of Nanomedicine. ACS NANO 2017; 11:2313-2381. [PMID: 28290206 PMCID: PMC5371978 DOI: 10.1021/acsnano.6b06040] [Citation(s) in RCA: 784] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Indexed: 04/14/2023]
Abstract
The design and use of materials in the nanoscale size range for addressing medical and health-related issues continues to receive increasing interest. Research in nanomedicine spans a multitude of areas, including drug delivery, vaccine development, antibacterial, diagnosis and imaging tools, wearable devices, implants, high-throughput screening platforms, etc. using biological, nonbiological, biomimetic, or hybrid materials. Many of these developments are starting to be translated into viable clinical products. Here, we provide an overview of recent developments in nanomedicine and highlight the current challenges and upcoming opportunities for the field and translation to the clinic.
Collapse
Affiliation(s)
- Beatriz Pelaz
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Christoph Alexiou
- ENT-Department, Section of Experimental Oncology & Nanomedicine
(SEON), Else Kröner-Fresenius-Stiftung-Professorship for Nanomedicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Ramon A. Alvarez-Puebla
- Department of Physical Chemistry, Universitat Rovira I Virgili, 43007 Tarragona, Spain
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Frauke Alves
- Department of Haematology and Medical Oncology, Department of Diagnostic
and Interventional Radiology, University
Medical Center Göttingen, 37075 Göttingen Germany
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - Anne M. Andrews
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Sumaira Ashraf
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Lajos P. Balogh
- AA Nanomedicine & Nanotechnology Consultants, North Andover, Massachusetts 01845, United States
| | - Laura Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy
| | - Alessandra Bestetti
- School of Chemistry & Bio21 Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Cornelia Brendel
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Susanna Bosi
- Department of Chemical
and Pharmaceutical Sciences, University
of Trieste, 34127 Trieste, Italy
| | - Monica Carril
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
- Ikerbasque, Basque Foundation
for Science, 48013 Bilbao, Spain
| | - Warren C. W. Chan
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Chunying Chen
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
| | - Xiaodong Chen
- School of Materials
Science and Engineering, Nanyang Technological
University, Singapore 639798
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine,
National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhen Cheng
- Molecular
Imaging Program at Stanford and Bio-X Program, Canary Center at Stanford
for Cancer Early Detection, Stanford University, Stanford, California 94305, United States
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Department of Instrument
Science and Engineering, School of Electronic Information and Electronical
Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials
Science and Engineering, Tongji University, Shanghai, China
| | - Christian Dullin
- Department of Haematology and Medical Oncology, Department of Diagnostic
and Interventional Radiology, University
Medical Center Göttingen, 37075 Göttingen Germany
| | - Alberto Escudero
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
- Instituto
de Ciencia de Materiales de Sevilla. CSIC, Universidad de Sevilla, 41092 Seville, Spain
| | - Neus Feliu
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Mingyuan Gao
- Institute of Chemistry, Chinese
Academy of Sciences, 100190 Beijing, China
| | | | - Yury Gogotsi
- Department of Materials Science and Engineering and A.J. Drexel Nanomaterials
Institute, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Arnold Grünweller
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Zhongwei Gu
- College of Polymer Science and Engineering, Sichuan University, 610000 Chengdu, China
| | - Naomi J. Halas
- Departments of Physics and Astronomy, Rice
University, Houston, Texas 77005, United
States
| | - Norbert Hampp
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Roland K. Hartmann
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Mark C. Hersam
- Departments of Materials Science and Engineering, Chemistry,
and Medicine, Northwestern University, Evanston, Illinois 60208, United States
| | - Patrick Hunziker
- University Hospital, 4056 Basel, Switzerland
- CLINAM,
European Foundation for Clinical Nanomedicine, 4058 Basel, Switzerland
| | - Ji Jian
- Department of Polymer Science and Engineering and Center for
Bionanoengineering and Department of Chemical and Biological Engineering, Zhejiang University, 310027 Hangzhou, China
| | - Xingyu Jiang
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
| | - Philipp Jungebluth
- Thoraxklinik Heidelberg, Universitätsklinikum
Heidelberg, 69120 Heidelberg, Germany
| | - Pranav Kadhiresan
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | | | | | - Jindřich Kopeček
- Biomedical Polymers Laboratory, University of Utah, Salt Lake City, Utah 84112, United States
| | - Nicholas A. Kotov
- Emergency Medicine, University of Michigan, Ann Arbor, Michigan 48019, United States
| | - Harald F. Krug
- EMPA, Federal Institute for Materials
Science and Technology, CH-9014 St. Gallen, Switzerland
| | - Dong Soo Lee
- Department of Molecular Medicine and Biopharmaceutical
Sciences and School of Chemical and Biological Engineering, Seoul National University, Seoul, South Korea
| | - Claus-Michael Lehr
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- HIPS - Helmhotz Institute for Pharmaceutical Research Saarland, Helmholtz-Center for Infection Research, 66123 Saarbrücken, Germany
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS), 100190 Beijing, China
| | - Mei Ling Lim
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Luis M. Liz-Marzán
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
- Ikerbasque, Basque Foundation
for Science, 48013 Bilbao, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine, Ciber-BBN, 20014 Donostia - San Sebastián, Spain
| | - Xiaowei Ma
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS), 100190 Beijing, China
| | - Paolo Macchiarini
- Laboratory of Bioengineering Regenerative Medicine (BioReM), Kazan Federal University, 420008 Kazan, Russia
| | - Huan Meng
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Helmuth Möhwald
- Department of Interfaces, Max-Planck
Institute of Colloids and Interfaces, 14476 Potsdam, Germany
| | - Paul Mulvaney
- School of Chemistry & Bio21 Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andre E. Nel
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Shuming Nie
- Emory University, Atlanta, Georgia 30322, United States
| | - Peter Nordlander
- Departments of Physics and Astronomy, Rice
University, Houston, Texas 77005, United
States
| | - Teruo Okano
- Tokyo Women’s Medical University, Tokyo 162-8666, Japan
| | | | - Tai Hyun Park
- Department of Molecular Medicine and Biopharmaceutical
Sciences and School of Chemical and Biological Engineering, Seoul National University, Seoul, South Korea
- Advanced Institutes of Convergence Technology, Suwon, South Korea
| | - Reginald M. Penner
- Department of Chemistry, University of
California, Irvine, California 92697, United States
| | - Maurizio Prato
- Department of Chemical
and Pharmaceutical Sciences, University
of Trieste, 34127 Trieste, Italy
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
- Ikerbasque, Basque Foundation
for Science, 48013 Bilbao, Spain
| | - Victor Puntes
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
- Institut Català de Nanotecnologia, UAB, 08193 Barcelona, Spain
- Vall d’Hebron University Hospital
Institute of Research, 08035 Barcelona, Spain
| | - Vincent M. Rotello
- Department
of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Amila Samarakoon
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Raymond E. Schaak
- Department of Chemistry, The
Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Youqing Shen
- Department of Polymer Science and Engineering and Center for
Bionanoengineering and Department of Chemical and Biological Engineering, Zhejiang University, 310027 Hangzhou, China
| | - Sebastian Sjöqvist
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Andre G. Skirtach
- Department of Interfaces, Max-Planck
Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Department of Molecular Biotechnology, University of Ghent, B-9000 Ghent, Belgium
| | - Mahmoud G. Soliman
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Molly M. Stevens
- Department of Materials,
Department of Bioengineering, Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Institute of Biomedical
Engineering, National Tsing Hua University, Hsinchu City, Taiwan,
ROC 300
| | - Ben Zhong Tang
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Hong Kong, China
| | - Rainer Tietze
- ENT-Department, Section of Experimental Oncology & Nanomedicine
(SEON), Else Kröner-Fresenius-Stiftung-Professorship for Nanomedicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Buddhisha N. Udugama
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - J. Scott VanEpps
- Emergency Medicine, University of Michigan, Ann Arbor, Michigan 48019, United States
| | - Tanja Weil
- Institut für
Organische Chemie, Universität Ulm, 89081 Ulm, Germany
- Max-Planck-Institute for Polymer Research, 55128 Mainz, Germany
| | - Paul S. Weiss
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Itamar Willner
- Institute of Chemistry, The Center for
Nanoscience and Nanotechnology, The Hebrew
University of Jerusalem, Jerusalem 91904, Israel
| | - Yuzhou Wu
- Max-Planck-Institute for Polymer Research, 55128 Mainz, Germany
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 430074 Wuhan, China
| | | | - Zhao Yue
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Qian Zhang
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Qiang Zhang
- School of Pharmaceutical Science, Peking University, 100191 Beijing, China
| | - Xian-En Zhang
- National Laboratory of Biomacromolecules,
CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
| | - Xin Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Wolfgang J. Parak
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
| |
Collapse
|
24
|
Shen S, Liu M, Li T, Lin S, Mo R. Recent progress in nanomedicine-based combination cancer therapy using a site-specific co-delivery strategy. Biomater Sci 2017; 5:1367-1381. [DOI: 10.1039/c7bm00297a] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review article highlights the recent progresses in nanomedicine-based combination cancer therapy via site-specific co-delivery strategies.
Collapse
Affiliation(s)
- Shiyang Shen
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Meng Liu
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Teng Li
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Shiqi Lin
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Ran Mo
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| |
Collapse
|
25
|
Mu CF, Xiong Y, Bai X, Sheng YJ, Cui J. Codelivery of Ponatinib and SAR302503 by Active Bone-Targeted Polymeric Micelles for the Treatment of Therapy-Resistant Chronic Myeloid Leukemia. Mol Pharm 2016; 14:274-283. [PMID: 27957861 DOI: 10.1021/acs.molpharmaceut.6b00872] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Point mutations in the BCR-ABL1 domain and primitive chronic myelogenous leukemia (CML) cells existing in the bone marrow environment insensitive to tyrosine kinase inhibitors (TKIs) have become two major challenges in the CML therapy. In this study, combined TKI ponatinib and JAK2 inhibitor SAR302503 short-term treatment effectively suppressed growth and promoted apoptosis of BaF3/T315I cells in cytokine-containing medium in vitro. SAR302503 prevented cytokine-dependent resistance to ponatinib via inhibition of JAK2/STAT5 phosphorylation. Codelivery of ponatinib and SAR302503 by active bone-targeted polymeric micellar formulation greatly increased the drug accumulation in medullary cavity. The therapeutic efficacy of bone-targeted formulation was demonstrated in BaF3/T315I cells inoculated murine model with no dose-limited toxicity detectable in health mice. Thus, the intravenous injectable bone-homing ponatinib and SAR302503 micellar formulation represents a promising strategy for the treatment of therapy-resistant CML.
Collapse
Affiliation(s)
- Chao-Feng Mu
- Department of Pharmaceutics, College of Pharmacy, Zhejiang Chinese Medical University , Hangzhou, Zhejiang 310053, China
| | - Yang Xiong
- Department of Pharmaceutics, College of Pharmacy, Zhejiang Chinese Medical University , Hangzhou, Zhejiang 310053, China
| | - Xue Bai
- Department of Pharmaceutics, College of Pharmacy, Zhejiang Chinese Medical University , Hangzhou, Zhejiang 310053, China
| | - Yun-Jie Sheng
- Department of Pharmaceutics, College of Pharmacy, Zhejiang Chinese Medical University , Hangzhou, Zhejiang 310053, China
| | - Jiajun Cui
- Department of Biochemistry, College of Medicine, Yichun University , Yichun, Jiangxi 336000, China
| |
Collapse
|
26
|
|
27
|
Shi S, Zhou M, Li X, Hu M, Li C, Li M, Sheng F, Li Z, Wu G, Luo M, Cui H, Li Z, Fu R, Xiang M, Xu J, Zhang Q, Lu L. Synergistic active targeting of dually integrin αvβ3/CD44-targeted nanoparticles to B16F10 tumors located at different sites of mouse bodies. J Control Release 2016; 235:1-13. [PMID: 27235150 DOI: 10.1016/j.jconrel.2016.05.050] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 05/13/2016] [Accepted: 05/23/2016] [Indexed: 12/31/2022]
Abstract
Conventional enhanced permeation and retention (EPR) mediates the effects of many drugs, including the accumulation of nanocarriers at tumor sites, but its efficiency remains low. In this study, this limitation was overcome by developing a dual-targeting delivery system based on hyaluronan (HA, a major ligand of CD44) and tetraiodothyroacetic acid (tetrac, a specific ligand of αvβ3), which was exploited to carry docetaxel (DTX) for the synergistic active targeting to tumors. First, a tetrac-HA (TeHA) conjugate was synthesized and grafted onto the surfaces of solid lipid nanoparticles (SLNs) (TeHA-SLNs/DTX), with a high encapsulation efficiency of >91.6%. The resulting SLNs exhibited an approximately toroid morphology revealed using TEM. The cellular uptake and cytotoxicity of various formulations on CD44/αvβ3-enriched B16F10 cells were then assessed, and both results confirmed the selective uptake and high cytotoxicity of the TeHA-SLNs/DTX in a TeHA-dependent manner. In vivo imaging and vessel distribution tests revealed the efficiency of synergistic active targeting was higher than that of EPR-mediated passive targeting by the TeHA-SLNs to αvβ3-expressing tumor blood vessels and CD44-expressing tumor cells via selective targeting. Finally, in both xenograft tumor mice and in situ lung metastasis tumor mice, tumor growth was significantly inhibited by TeHA-SLNs/DTX. Therefore, TeHA-SLNs are an efficient system for the dual-targeted delivery of drugs to treat cancer in vivo.
Collapse
Affiliation(s)
- Sanjun Shi
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China.
| | - Min Zhou
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Xin Li
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Min Hu
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Chenwen Li
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Min Li
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Fangfang Sheng
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Zhuoheng Li
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Guolin Wu
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Minghe Luo
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Huanhuan Cui
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Ziwei Li
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Ruoqiu Fu
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Mingfeng Xiang
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Jing Xu
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Qian Zhang
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Laichun Lu
- Department of Pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China.
| |
Collapse
|
28
|
Goins B, Phillips WT, Bao A. Strategies for improving the intratumoral distribution of liposomal drugs in cancer therapy. Expert Opin Drug Deliv 2016; 13:873-89. [PMID: 26981891 DOI: 10.1517/17425247.2016.1167035] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION A major limitation of current liposomal cancer therapies is the inability of liposome therapeutics to penetrate throughout the entire tumor mass. This inhomogeneous distribution of liposome therapeutics within the tumor has been linked to treatment failure and drug resistance. Both liposome particle transport properties and tumor microenvironment characteristics contribute to this challenge in cancer therapy. This limitation is relevant to both intravenously and intratumorally administered liposome therapeutics. AREAS COVERED Strategies to improve the intratumoral distribution of liposome therapeutics are described. Combination therapies of intravenous liposome therapeutics with pharmacologic agents modulating abnormal tumor vasculature, interstitial fluid pressure, extracellular matrix components, and tumor associated macrophages are discussed. Combination therapies using external stimuli (hyperthermia, radiofrequency ablation, magnetic field, radiation, and ultrasound) with intravenous liposome therapeutics are discussed. Intratumoral convection-enhanced delivery (CED) of liposomal therapeutics is reviewed. EXPERT OPINION Optimization of the combination therapies and drug delivery protocols are necessary. Further research should be conducted in appropriate cancer types with consideration of physiochemical features of liposomes and their timing sequence. More investigation of the role of tumor associated macrophages in intratumoral distribution is warranted. Intratumoral infusion of liposomes using CED is a promising approach to improve their distribution within the tumor mass.
Collapse
Affiliation(s)
- Beth Goins
- a Department of Radiology , University of Texas Health Science Center San Antonio , San Antonio , TX , USA
| | - William T Phillips
- a Department of Radiology , University of Texas Health Science Center San Antonio , San Antonio , TX , USA
| | - Ande Bao
- b Department of Radiation Oncology, School of Medicine, Case Western Reserve University/University Hospitals Case Medical Center , Cleveland , OH , USA
| |
Collapse
|
29
|
Yang HY, Jang MS, Gao GH, Lee JH, Lee DS. Construction of redox/pH dual stimuli-responsive PEGylated polymeric micelles for intracellular doxorubicin delivery in liver cancer. Polym Chem 2016. [DOI: 10.1039/c5py01808k] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A schematic of the complete process of DOX-loaded mPEG-SS-PNLG micelles uptake into tumor cells and intracellular release of DOX; size distribution of mPEG-SS-PNLG (90%) micelles and in vivo antitumor efficacy.
Collapse
Affiliation(s)
- Hong Yu Yang
- Theranostic Macromolecules Research Center
- School of Chemical Engineering
- Sungkyunkwan University
- Suwon 440-746
- Republic of Korea
| | - Moon-Sun Jang
- Department of Radiology
- Samsung Medical Center
- Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging
- Samsung Biomedical Research Institute
- Seoul 135-710
| | - Guang Hui Gao
- Engineering Research Center of Synthetic Resin and Special Fiber
- Ministry of Education
- Changchun University of Technology
- Changchun 130012
- China
| | - Jung Hee Lee
- Department of Radiology
- Samsung Medical Center
- Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging
- Samsung Biomedical Research Institute
- Seoul 135-710
| | - Doo Sung Lee
- Theranostic Macromolecules Research Center
- School of Chemical Engineering
- Sungkyunkwan University
- Suwon 440-746
- Republic of Korea
| |
Collapse
|
30
|
Liu H, Xie Y, Zhang Y, Cai Y, Li B, Mao H, Yu R. CA4-loaded doxorubicin prodrug coating Fe3O4 nanoparticles for tumor combination therapy. RSC Adv 2016. [DOI: 10.1039/c6ra24562e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Fe3O4 nanoparticles (NPs) have attracted a great deal of attention due to their magnetic properties, low toxicity, high surface area and their small sizes.
Collapse
Affiliation(s)
- Hongmei Liu
- Brain Hospital
- Affiliated Hospital of Xuzhou Medical University
- Xuzhou
- China
- Insititute of Nervous System Diseases
| | - Yandong Xie
- Brain Hospital
- Affiliated Hospital of Xuzhou Medical University
- Xuzhou
- China
- Insititute of Nervous System Diseases
| | - Yafei Zhang
- Brain Hospital
- Affiliated Hospital of Xuzhou Medical University
- Xuzhou
- China
- Insititute of Nervous System Diseases
| | - Yifan Cai
- Brain Hospital
- Affiliated Hospital of Xuzhou Medical University
- Xuzhou
- China
- Insititute of Nervous System Diseases
| | - Baiyang Li
- Brain Hospital
- Affiliated Hospital of Xuzhou Medical University
- Xuzhou
- China
- Insititute of Nervous System Diseases
| | - Honglin Mao
- Brain Hospital
- Affiliated Hospital of Xuzhou Medical University
- Xuzhou
- China
- Insititute of Nervous System Diseases
| | - Rutong Yu
- Brain Hospital
- Affiliated Hospital of Xuzhou Medical University
- Xuzhou
- China
- Insititute of Nervous System Diseases
| |
Collapse
|
31
|
WITHDRAWN: Polymer assembly: Promising carriers as co-delivery systems for cancer therapy. Prog Polym Sci 2015. [DOI: 10.1016/j.progpolymsci.2015.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
32
|
Poojari R, Kini S, Srivastava R, Panda D. A Chimeric Cetuximab-Functionalized Corona as a Potent Delivery System for Microtubule-Destabilizing Nanocomplexes to Hepatocellular Carcinoma Cells: A Focus on EGFR and Tubulin Intracellular Dynamics. Mol Pharm 2015; 12:3908-23. [PMID: 26426829 DOI: 10.1021/acs.molpharmaceut.5b00337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Radhika Poojari
- Department of Biosciences
and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sudarshan Kini
- Department of Biosciences
and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Rohit Srivastava
- Department of Biosciences
and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Dulal Panda
- Department of Biosciences
and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
33
|
Micellar carriers for the delivery of multiple therapeutic agents. Colloids Surf B Biointerfaces 2015; 135:291-308. [PMID: 26263217 DOI: 10.1016/j.colsurfb.2015.07.046] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 07/16/2015] [Accepted: 07/19/2015] [Indexed: 12/27/2022]
Abstract
Multi-drug therapy is described as a simultaneous or sequential administration of two or more drugs with similar or different mechanisms of action and is recognized as a more efficient solution to combat successfully, various ailments. Polymeric micelles (PMs) are self-assemblies of block copolymers providing numerous opportunities for drug delivery. To date various micellar formulations were studied for delivery of drugs, nutraceuticals and genes; a few of them are in clinical trials. It was observed that there is an immense need for the development of PMs embedding multiple therapeutic agents to combat various ailments, including cancers, HIV/AIDS, malaria, multiple sclerosis, hypertension, infectious diseases, cardiovascular and metabolic diseases, immune disorders and many psychiatric disorders. Several combinations of drug-drug, drug-nutraceutical, drug-gene and drug-siRNA explored to date are detailed in this review, with a special emphasis on their potential and future perspectives. A summary of various preparation methods, characterization techniques and applications of PMs are also provided. This review presents a holistic approach on multi-drug delivery using micellar carriers and emphasizes on the development of therapeutic hybrids embedding novel combinations for safer and effective therapy.
Collapse
|
34
|
Dong Y, Yang J, Liu H, Wang T, Tang S, Zhang J, Zhang X. Site-Specific Drug-Releasing Polypeptide Nanocarriers Based on Dual-pH Response for Enhanced Therapeutic Efficacy against Drug-Resistant Tumors. Theranostics 2015; 5:890-904. [PMID: 26000060 PMCID: PMC4440445 DOI: 10.7150/thno.11821] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 03/31/2015] [Indexed: 11/05/2022] Open
Abstract
To enhance effective drug accumulation in drug-resistant tumors, a site-specific drug-releasing polypeptide system (PEG-Phis/Pasp-DOX/CA4) was exploited in response to tumor extracellular and intracellular pH. This system could firstly release the embedded tumor vascular inhibitor (CA4) to transiently 'normalize' vasculature and facilitate drug internalization to tumors efficiently, and then initiate the secondary pH-response to set the conjugated active anticancer drug (DOX) free in tumor cells. The encapsulated system (PEG-Phis/DOX/CA4), both CA4 and DOX embedding in the nanoparticles, was used as a control. Comparing with PEG-Phis/DOX/CA4, PEG-Phis/Pasp-DOX/CA4 exhibited enhanced cytotoxicity against DOX-sensitive and DOX-resistant cells (MCF-7 and MCF-7/ADR). Moreover, PEG-Phis/Pasp-DOX/CA4 resulted in enhanced therapeutic efficacy in drug-resistant tumors with reduced toxicity. These results suggested that this site-specific drug-releasing system could be exploited as a promising treatment for cancers with repeated administration.
Collapse
Affiliation(s)
- Yaqiong Dong
- 1. National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- 2. College of Chemistry & Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Jun Yang
- 1. National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Hongmei Liu
- 1. National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- 3. University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianyou Wang
- 4. Capital Institute of Pediatrics, Beijing, 100020, China
| | - Suoqin Tang
- 5. Department of Pediatrics, The General Hospital of People's Liberation Army, Beijing, 100853, China
| | - Jinchao Zhang
- 2. College of Chemistry & Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Xin Zhang
- 1. National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
35
|
Liu H, Zhang R, Niu Y, Li Y, Qiao C, Weng J, Li J, Zhang X, Xiao Z, Zhang X. Development of hypoxia-triggered prodrug micelles as doxorubicin carriers for tumor therapy. RSC Adv 2015. [DOI: 10.1039/c4ra14875d] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hypoxia-responsive prodrug micelles to deliver anticancer drug, which can selectively release the drugs to treat hypoxic tumor cells.
Collapse
|
36
|
Recent advances in targeted nanoparticles drug delivery to melanoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 11:769-94. [PMID: 25555352 DOI: 10.1016/j.nano.2014.11.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/31/2014] [Accepted: 11/15/2014] [Indexed: 12/30/2022]
Abstract
Melanoma is one of the most aggressive skin cancers, notorious for its high multidrug resistance and low survival rate. Conventional therapies (e.g., dacarbazine, interferon-alpha-2b and interleukin-2) are limited by low response rate and demonstrate no overall survival benefit. Novel targeted therapies (e.g., vemurafenib, dabrafenib and trametinib) have higher initial response rate and clear impact on the overall survival, but relapse usually occurs within 6 to 9 months. Although immunotherapy (e.g., ipilimumab, pembrolizumab and nivolumab) can achieve long-term and durable response, rate of adverse events is extremely high. With the development of nanotechnology, the applications of nanocarriers are widely expected to change the landscape of melanoma therapy for foreseeable future. In this review, we will relate recent advances in the application of multifunctional nanocarriers for targeted drug delivery to melanoma, in melanoma nanotheranostics and combination therapy, and nanopharmaceutical associated melanoma clinical trials, followed by challenges and perspectives. From the clinical editor: The team of authors describes the current treatment regimes of malignant melanoma emphasizing the importance of achieving a better efficacy and the need to develop a better understanding of melanoma tumorigenesis.
Collapse
|
37
|
Wang X, Li S, Shi Y, Chuan X, Li J, Zhong T, Zhang H, Dai W, He B, Zhang Q. The development of site-specific drug delivery nanocarriers based on receptor mediation. J Control Release 2014; 193:139-53. [DOI: 10.1016/j.jconrel.2014.05.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/13/2014] [Accepted: 05/17/2014] [Indexed: 01/28/2023]
|
38
|
Chuan X, Song Q, Lin J, Chen X, Zhang H, Dai W, He B, Wang X, Zhang Q. Novel free-paclitaxel-loaded redox-responsive nanoparticles based on a disulfide-linked poly(ethylene glycol)-drug conjugate for intracellular drug delivery: synthesis, characterization, and antitumor activity in vitro and in vivo. Mol Pharm 2014; 11:3656-70. [PMID: 25208098 DOI: 10.1021/mp500399j] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
To address the obstacles facing cancer chemotherapeutics, including toxicity, side effects, water insolubility, and lack of tumor selectivity, a novel stimuli-responsive drug-delivery system was developed based on paclitaxel-loaded poly(ethylene glycol)-disulfide-paclitaxel conjugate nanoparticles (PEG-SS-PTX/PTX NPs). The formulation emphasizes several benefits, including polymer-drug conjugates/prodrugs, self-assembled NPs, high drug content, redox responsiveness, and programmed drug release. The PTX-loaded, self-assembled NPs, with a uniform size of 103 nm, characterized by DLS, TEM, XRD, DSC, and (1)H NMR, exhibited excellent drug-loading capacity (15.7%) and entrapment efficiency (93.3%). PEG-SS-PTX/PTX NPs were relatively stable under normal conditions but disassembled quickly under reductive conditions, as indicated by their triggered-aggregation phenomena and drug-release profile in the presence of dithiothreitol (DTT), a reducing agent. Additionally, by taking advantage of the difference in the drug-release rates between physically loaded and chemically conjugated drugs, a programmed drug-release phenomenon was observed, which was attributed to a higher concentration and longer action time of the drugs. The influence of PEG-SS-PTX/PTX NPs on in vitro cytotoxicity, cell cycle progression, and cellular apoptosis was determined in the MCF-7 cell line, and the NPs demonstrated a superior anti-proliferative activity associated with PTX-induced cell cycle arrest in G2/M phase and apoptosis compared to their nonresponsive counterparts. Moreover, the redox-responsive NPs were more efficacious than both free PTX and the non-redox-responsive formulation at equivalent doses of PTX in a breast cancer xenograft mouse model. This redox-responsive PTX drug delivery system is promising and can be explored for use in effective intracellular drug delivery.
Collapse
Affiliation(s)
- Xingxing Chuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University , Beijing 100191, China
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Li J, Li Z, Li M, Zhang H, Xie Z. Synergistic effect and drug-resistance relief of paclitaxel and cisplatin caused by Co-delivery using polymeric micelles. J Appl Polym Sci 2014. [DOI: 10.1002/app.41440] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Jing Li
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry, Chinese Academy of Sciences; Changchun 130022 People's Republic of China
| | - Zhihong Li
- Department of Thoracic Surgery; The First Hospital of Jilin University; Changchun 130021 People's Republic of China
| | - Minghe Li
- Department of Oral and Maxillofacial Surgery; School of Stomatology Hospital of Jilin University; Changchun 130021 People's Republic of China
| | - Hong Zhang
- Department of Thoracic Surgery; The First Hospital of Jilin University; Changchun 130021 People's Republic of China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry, Chinese Academy of Sciences; Changchun 130022 People's Republic of China
| |
Collapse
|
40
|
Luo X, Zhang H, Chen M, Wei J, Zhang Y, Li X. Antimetastasis and antitumor efficacy promoted by sequential release of vascular disrupting and chemotherapeutic agents from electrospun fibers. Int J Pharm 2014; 475:438-49. [PMID: 25218185 DOI: 10.1016/j.ijpharm.2014.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/17/2014] [Accepted: 09/06/2014] [Indexed: 01/15/2023]
Abstract
The vasculature in tumor microenvironment plays important roles in the tumor growth and metastasis, and the combination of vascular disrupting agents with chemotherapeutic drugs should be effective in inhibiting tumor progression. But the dosing schedules are essential to achieve a balance between vascular collapse and intratumoral uptake of chemotherapeutic agents. In the current study, emulsion and blend electrospinning were used to create compartmental fibers accommodating both combretastatin A-4 (CA4) and hydroxycamptothecin (HCPT). The release durations of CA4 and HCPT were modulated through the structure of fibers for dual drug loadings and the inoculation of 2-hydroxypropyl-β-cyclodextrin in fiber matrices. Under a noncontact cell coculture in Transwell, the sequential release of CA4 and HCPT indicated a sequential killing of endothelial and tumor cells. In an orthotopic breast tumor model, all the CA4/HCPT-loaded fibers showed superior antitumor efficacy and higher survival rate than fibers with loaded individual drug. Compared with fibrous mats with infiltrated free CA4 and fibers with extended release of CA4 for over 30 days, fibers with sustained release of CA4 for 3-7 days from CA4/HCPT-loaded fibers resulted in the most significant antitumor efficacy, tumor vasculature destruction, and the least tumor metastasis to lungs. A judicious selection of CA4 release durations in the combination therapy should be essential to enhance the tumor suppression efficacy and antimetastasis activity.
Collapse
Affiliation(s)
- Xiaoming Luo
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Hong Zhang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Maohua Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Jiaojun Wei
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Yun Zhang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China
| | - Xiaohong Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, PR China.
| |
Collapse
|
41
|
Lu J, Chuan X, Zhang H, Dai W, Wang X, Wang X, Zhang Q. Free paclitaxel loaded PEGylated-paclitaxel nanoparticles: preparation and comparison with other paclitaxel systems in vitro and in vivo. Int J Pharm 2014; 471:525-35. [PMID: 24858391 DOI: 10.1016/j.ijpharm.2014.05.032] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/01/2014] [Accepted: 05/18/2014] [Indexed: 11/19/2022]
Abstract
Previously, PEGylated paclitaxel (PEG-PTX) was found not favorable as a polymer prodrug because of its poor antitumor efficiency. But surprisingly, it was found in our study that PEG-PTX could form a novel nanoparticle system with free PTX. To address how this system works, we compared PTX loaded PEG-PTX nanoparticles (PEG-PTX/PTX) with PTX loaded PEG-PLA micelles (PEG-PLA/PTX) or PTX injection available (Taxol(®)) in vitro and in vivo. Firstly, it was found that PEG-PTX/PTX was more stable in aqueous solution than PEG-PLA/PTX in terms of PTX crystal formation and drug release. Then it was demonstrated that coumarin loaded PEG-PTX nanoparticles had a much higher uptake in MCF-7 cells compared to coumarin loaded PEG-PLA micelles. The in vivo imaging study revealed that DIR or DID (near infrared fluorescent substances) loaded PEG-PTX nanoparticles distributed more in tumors in MCF-7 tumor bearing mice than DIR or DID loaded PEG-PLA micelles and solvent system of Taxol(®). In the efficacy study with MCF-7 tumor bearing mice, PEG-PTX/PTX showed significantly higher antitumor activity than PEG-PLA/PTX at the same PTX dosage. At the dose of 10mg free PTX per kg, PEG-PTX/PTX displayed similar efficacy as Taxol(®) but less toxicity evaluated by the loss of body weight. With the increase of free PTX to 15 mg/kg, PEG-PTX/PTX showed significantly better efficacy than Taxol(®). In conclusion, with favorable characteristics in stability, cellular uptake, cytotoxicity, biodistribution, safety and efficacy, PEG-PTX/PTX seems highly potential as a nanocarrier for PTX delivery.
Collapse
Affiliation(s)
- Jingkai Lu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xingxing Chuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hua Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenbing Dai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xinglin Wang
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin 300193, China
| | - Xueqing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
42
|
Feng Q, Yu MZ, Wang JC, Hou WJ, Gao LY, Ma XF, Pei XW, Niu YJ, Liu XY, Qiu C, Pang WH, Du LL, Zhang Q. Synergistic inhibition of breast cancer by co-delivery of VEGF siRNA and paclitaxel via vapreotide-modified core-shell nanoparticles. Biomaterials 2014; 35:5028-38. [PMID: 24680191 DOI: 10.1016/j.biomaterials.2014.03.012] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/07/2014] [Indexed: 01/05/2023]
Abstract
A somatostatin analog, vapreotide (VAP), can be used as a ligand for targeting drug delivery based on its high affinity to somatostatin receptors (SSTRs), which is overexpressed in many tumor cells. RNA interference plays an important role on downregulation of vascular endothelial growth factor (VEGF), which is important for tumor growth, progression and metastasis. To improve tumor therapy efficacy, the vapreotide-modified core-shell type nanoparticles co-encapsulating VEGF targeted siRNA (siVEGF) and paclitaxel (PTX), termed as VAP-PLPC/siRNA NPs, were developed in this study. When targeted via somatostatin receptors to tumor cells, the VAP-PLPC/siRNA NPs could simultaneously delivery siVEGF and PTX into cells and achieve a synergistic inhibition of tumor growth. Interestingly, in vitro cell uptake and gene silencing experiments demonstrated that the targeted VAP-PLPC/siRNA NPs exhibited significant higher intracellular siRNA accumulation and VEGF downregulation in human breast cancer MCF-7 cells, compared to those of the non-targeted PEG-PLPC/siRNA NPs. More importantly, in vivo results further demonstrated that the targeted VAP-PLPC/siRNA NPs had significant stronger drug distribution in tumor tissues and tumor growth inhibition efficacy via receptor-mediated targeting delivery, accompany with an obvious inhibition of neovascularization induced by siVEGF silencing. These results suggested that the co-delivery of siRNA and paclitaxel via vapreotide-modified core-shell nanoparticles would be a promising approach for tumor targeted therapy.
Collapse
Affiliation(s)
- Qiang Feng
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Min-Zhi Yu
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Jian-Cheng Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China.
| | - Wen-Jie Hou
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Ling-Yan Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Xiao-Fei Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Xi-Wei Pei
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Yu-Jie Niu
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Xiao-Yan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Chong Qiu
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Wen-Hao Pang
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Li-Li Du
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of pharmaceutical sciences, Peking University, Beijing 100191, PR China
| |
Collapse
|
43
|
Polypeptide-based combination of paclitaxel and cisplatin for enhanced chemotherapy efficacy and reduced side-effects. Acta Biomater 2014; 10:1392-402. [PMID: 24316362 DOI: 10.1016/j.actbio.2013.11.026] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 11/22/2013] [Accepted: 11/26/2013] [Indexed: 12/11/2022]
Abstract
A novel methoxy poly(ethylene glycol)-b-poly(l-glutamic acid)-b-poly(l-phenylalanine) (mPEG-b-P(Glu)-b-P(Phe)) triblock copolymer was prepared and explored as a micelle carrier for the co-delivery of paclitaxel (PTX) and cisplatin (cis-diamminedichlo-platinum, CDDP). PTX and CDDP were loaded inside the hydrophobic P(Phe) inner core and chelated to the middle P(Glu) shell, respectively, while mPEG provided the outer corona for prolonged circulation. An in vitro release profile of the PTX+CDDP-loaded micelles showed that the CDDP chelation cross-link prevented an initial burst release of PTX. The PTX+CDDP-loaded micelles exhibited a high synergism effect in the inhibition of A549 human lung cancer cell line proliferation over 72 h incubation. For the in vivo treatment of xenograft human lung tumor, the PTX+CDDP-loaded micelles displayed an obvious tumor inhibiting effect with a 83.1% tumor suppression rate (TSR%), which was significantly higher than that of a free drug combination or micelles with a single drug. In addition, more importantly, the enhanced anti-tumor efficacy of the PTX+CDDP-loaded micelles came with reduced side-effects. No obvious body weight loss occurred during the treatment of A549 tumor-bearing mice with the PTX+CDDP-loaded micelles. Thus, the polypeptide-based combination of PTX and CDDP may provide useful guidance for effective and safe cancer chemotherapy.
Collapse
|
44
|
Zhao L, Zhai G. Preparation and in vitro and in vivo evaluation of RGD modified curcumin loaded PEG-PLA micelles. J Control Release 2013. [DOI: 10.1016/j.jconrel.2013.08.252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
45
|
Yu KF, Zhang WQ, Luo LM, Song P, Li D, Du R, Ren W, Huang D, Lu WL, Zhang X, Zhang Q. The antitumor activity of a doxorubicin loaded, iRGD-modified sterically-stabilized liposome on B16-F10 melanoma cells: in vitro and in vivo evaluation. Int J Nanomedicine 2013; 8:2473-85. [PMID: 23885174 PMCID: PMC3716561 DOI: 10.2147/ijn.s46962] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Considering the fact that iRGD (tumor-homing peptide) demonstrates tumor-targeting and tumor-penetrating activity, and that B16-F10 (murine melanoma) cells overexpress both αv integrin receptor and neuropilin-1 (NRP-1), the purpose of this study was to prepare a novel doxorubicin (DOX)-loaded, iRGD-modified, sterically-stabilized liposome (SSL) (iRGD-SSL-DOX) in order to evaluate its antitumor activity on B16-F10 melanoma cells in vitro and in vivo. The iRGD-SSL-DOX was prepared using a thin-film hydration method. The characteristics of iRGD-SSL-DOX were evaluated. The in vitro leakage of DOX from iRGD-SSL-DOX was tested. The in vitro tumor-targeting and tumor-penetrating characteristics of iRGD-modified liposomes on B16-F10 cells were investigated. The in vivo tumor-targeting and tumor-penetrating activities of iRGD-modified liposomes were performed in B16-F10 tumor-bearing nude mice. The antitumor effect of iRGD-SSL-DOX was evaluated in B16-F10 tumor-bearing C57BL/6 mice in vivo. The average particle size of the iRGD-SSL-DOX was found to be 91 nm with a polydispersity index (PDI) of 0.16. The entrapment efficiency of iRGD-SSL-DOX was 98.36%. The leakage of DOX from iRGD-SSL-DOX at the 24-hour time point was only 7.5%. The results obtained from the in vitro flow cytometry and confocal microscopy, as well as in vivo biodistribution and confocal immunofluorescence microscopy experiments, indicate that the tumor-targeting and tumor-penetrating activity of the iRGD-modified SSL was higher than that of unmodified SSL. In vivo antitumor activity results showed that the antitumor effect of iRGD-SSL-DOX against melanoma tumors was higher than that of SSL-DOX in B16-F10 tumor-bearing mice. In conclusion, the iRGD-SSL-DOX is a tumor-targeting and tumor-penetrating peptide modified liposome which has significant antitumor activity against melanoma tumors.
Collapse
Affiliation(s)
- Ke-Fu Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Tao Y, Han J, Wang X, Dou H. Nano-formulation of paclitaxel by vitamin E succinate functionalized pluronic micelles for enhanced encapsulation, stability and cytotoxicity. Colloids Surf B Biointerfaces 2013; 102:604-10. [DOI: 10.1016/j.colsurfb.2012.08.062] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/10/2012] [Accepted: 08/31/2012] [Indexed: 01/25/2023]
|
47
|
A review on comb-shaped amphiphilic polymers for hydrophobic drug solubilization. Ther Deliv 2012; 3:59-79. [PMID: 22833933 DOI: 10.4155/tde.11.130] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Comb-shaped amphiphilic polymers are rapidly emerging as an alternative approach to amphiphilic block copolymers for hydrophobic drug solubilization. These polymers consist of a homopolymer or copolymer backbone to which hydrophobic and hydrophilic pendant groups can be grafted resulting in a comb-like architecture. The hydrophobic pendants may consist of homopolymers, copolymers and other low-molecular weight hydrophobic structures. In this review, we focus on hydrophobically modified preformed homopolymers. Comb-shaped amphiphilic polymers possess reduced critical aggregation concentration values compared with traditional surfactant micelles indicating increased stability with decreased disruption experienced on dilution. They have been fabricated with diverse architectures and multifunctional properties such as site-specific targeting and external stimuli-responsive nature. The application of comb-shaped amphiphilic polymers is expanding; here we report on the progress achieved so far in hydrophobic drug solubilization for both intravenous and oral delivery.
Collapse
|
48
|
Ma M, Hao Y, Liu N, Yin Z, Wang L, Liang X, Zhang X. A novel lipid-based nanomicelle of docetaxel: evaluation of antitumor activity and biodistribution. Int J Nanomedicine 2012; 7:3389-98. [PMID: 22848167 PMCID: PMC3405881 DOI: 10.2147/ijn.s29827] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose A lipid-based, nanomicelle-loaded docetaxel (M-DOC) was designed and characterized. Optical imaging was employed to evaluate the pharmacokinetics and antitumor efficacy of docetaxel in vivo. Materials and methods The M-DOC was prepared using the emulsion-diffusion method. Transmission electron microscopy and dynamic light scattering were used to assess the morphology and particle size of the M-DOC. Critical micelle concentrations, their stability under physiological conditions, and their encapsulation efficiency – as measured by high-performance liquid chromatography – were assessed. Pharmacological features were evaluated in two different animal models by comparing M-DOC treatments with docetaxel injections (I-DOC). Bioluminescence imaging was used to assess antitumor activity and docetaxel distribution in vivo, using nude mice injected with luciferase-expressing MDA-MB-231 human breast tumor cells. In addition, animals injected with B16 melanoma cells were used to measure survival time and docetaxel distribution. Results The M-DOC was prepared as round, uniform spheres with an effective diameter of 20.8 nm. The critical micelle concentration of the original emulsion was 0.06%. Satisfactory encapsulation efficiency (87.6% ± 3.0%) and 12-hour stability were achieved. Xenograft results demonstrated that the M-DOC was more effective in inhibiting tumor growth, without significantly changing body weight. Survival was prolonged by 12.6% in the M-DOC group. Tumor growth inhibitory rates in the M-DOC and I-DOC groups were 91.2% and 57.8% in volume and 71.8% and 44.9% in weight, respectively. Optical bioluminescence imaging of tumor growths yielded similar results. Area under the curve(0–6 hour) levels of docetaxel in blood and tumors were significantly higher in the M-DOC group (15.9 ± 3.2 μg/mL−1, 601.1 ± 194.5 μg/g−1) than in the I-DOC group (7.2 ± 1.7 μg/mL−1, 357.8 ± 86.2 μg/g−1). The fluorescent dye 1,1-dioctadecyl-3,3,3,3′-tetramethylindotricarbocyanine iodide mimicked M-DOC in optical imaging, and accumulated more in tumors in comparison with I-DOC. Conclusion These results suggest that the lipid-based nanomicelle system was effective in inhibiting tumor growth, with little toxicity. Moreover, we have developed a noninvasive optical imaging method for antitumor drug evaluation, which merits further analysis for potential clinical applications.
Collapse
Affiliation(s)
- Mingshu Ma
- Department of Pharmacology and Pharmaceutical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Kulthe SS, Choudhari YM, Inamdar NN, Mourya V. Polymeric micelles: authoritative aspects for drug delivery. Des Monomers Polym 2012. [DOI: 10.1080/1385772x.2012.688328] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Affiliation(s)
- Sushant S. Kulthe
- a Government College of Pharmacy , Aurangabad , 431005 , Maharashtra , India
| | - Yogesh M. Choudhari
- a Government College of Pharmacy , Aurangabad , 431005 , Maharashtra , India
| | - Nazma N. Inamdar
- a Government College of Pharmacy , Aurangabad , 431005 , Maharashtra , India
| | - Vishnukant Mourya
- a Government College of Pharmacy , Aurangabad , 431005 , Maharashtra , India
| |
Collapse
|
50
|
Zhao ZX, Gao SY, Wang JC, Chen CJ, Zhao EY, Hou WJ, Feng Q, Gao LY, Liu XY, Zhang LR, Zhang Q. Self-assembly nanomicelles based on cationic mPEG-PLA-b-Polyarginine(R15) triblock copolymer for siRNA delivery. Biomaterials 2012; 33:6793-807. [PMID: 22721724 DOI: 10.1016/j.biomaterials.2012.05.067] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 05/29/2012] [Indexed: 12/25/2022]
Abstract
Due to the absence of safe and effective carriers for in vivo delivery, the applications of small interference RNA (siRNA) in clinic for therapeutic purposes have been limited. In this study, a biodegradable amphiphilic tri-block copolymer (mPEG(2000)-PLA(3000)-b-R(15)) composed of monomethoxy poly(ethylene glycol), poly(d,l-lactide) and polyarginine was synthesized and further self-assembled to cationic polymeric nanomicelles for in vivo siRNA delivery, with an average diameter of 54.30 ± 3.48 nm and a zeta potential of approximately 34.8 ± 1.77 mV. The chemical structures of the copolymers were well characterized by (1)H NMR spectroscopy and FT-IR spectra. In vitro cytotoxicity and hemolysis assays demonstrated that the polymeric nanomicelles showed greater cell viability and haemocompatibility than those of polyethyleneimine (PEI) or R(15) peptide. In vitro experiments demonstrated that EGFR targeted siRNA formulated in micelleplexes exhibited approximately 65% inhibition of EGFR expression on MCF-7 cells in a sequence-specific manner, which was comparable to Lipofectamine™ 2000. The results of intravenous administration showed Micelleplex/EGFR-siRNA significantly inhibited tumor growth in nude mice xenografted MCF-7 tumors, with a remarkable inhibition of EGFR expression. Furthermore, no positive activation of the innate immune responses and no significant body weight loss was observed during treatment suggested that this polymeric micelle delivery system is non-toxic. In conclusion, the present nanomicelles based on cationic mPEG(2000)-PLA(3000)-b-R(15) copolymer would be a safe and efficient nanocarrier for in vivo delivery of therapeutic siRNA.
Collapse
Affiliation(s)
- Zhi-Xia Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|