1
|
Gupta C, Jaipuria A, Gupta N. Inhalable Formulations to Treat Non-Small Cell Lung Cancer (NSCLC): Recent Therapies and Developments. Pharmaceutics 2022; 15:139. [PMID: 36678768 PMCID: PMC9861595 DOI: 10.3390/pharmaceutics15010139] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
Cancer has been the leading cause of mortalities, with lung cancer contributing 18% to overall deaths. Non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancers. The primary form of therapy used to treat lung cancer still includes oral and systemic administration of drugs, radiotherapy, or chemotherapy. Some patients have to go through a regime of combination therapy. Despite being the only available form of therapy, their use is limited due to the adverse effects, toxicity, and development of resistance over prolonged use. This led to a shift and progressive evolution into using pulmonary drug delivery systems. Being a non-invasive method of drug-administration and allowing localized delivery of drugs to cancer cells, inhalable drug delivery systems can lead to lower dosing and fewer systemic toxicities over other conventional routes. In this way, we can increase the actual local concentration of the drug in lungs, which will ultimately lead to better antitumor therapy. Nano-based systems also provide additional diagnostic advantages during lung cancer treatment, including imaging, screening, and tracking. Regardless of the advantages, pulmonary delivery is still in the early stages of development and various factors such as pharmacology, immunology, and toxicology should be taken into consideration for the development of suitable inhalable nano-based chemotherapeutic drugs. They face numerous physiological barriers such as lung retention and efficacy, and could also lead to toxicity due to prolonged exposure. Nano-carriers with a sustained drug release mechanism could help in overcoming these challenges. This review article will focus on the various inhalable formulations for targeted drug delivery, including nano-based delivery systems such as lipids, liposome, polymeric and inorganic nanocarriers, micelles, microparticles and nanoaggregates for lung cancer treatment. Various devices used in pulmonary drug delivery loaded on various nano-carriers are also discussed in detail.
Collapse
Affiliation(s)
- Chetna Gupta
- Department of Chemistry, Hansraj College, University of Delhi, Delhi 110007, India
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Aadya Jaipuria
- Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| | - Nikesh Gupta
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
2
|
Wu X, Tang T, Wei Y, Cummins KA, Wood DK, Pang H. Extracellular Vesicles Mediate the Intercellular Exchange of Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102441. [PMID: 35243822 PMCID: PMC8895114 DOI: 10.1002/advs.202102441] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 12/09/2021] [Indexed: 05/06/2023]
Abstract
To exert their therapeutic effects, nanoparticles (NPs) often need to travel into the tissues composed of multilayered cells. Accumulative evidence has revealed the crucial role of transcellular transport route (entry into one cell, exocytosis, and re-entry into another) in this process. While NP endocytosis and subcellular transport are intensively characterized, the exocytosis and re-entry steps are poorly understood, which becomes a barrier for NP delivery into complex tissues. Here, the authors term the exocytosis and re-entry steps together as intercellular exchange. A collagen-based three-dimension assay is developed to specifically quantify the intercellular exchange of NPs, and distinguish the contributions of several potential mechanisms. The authors show that NPs can be exocytosed freely or enclosed inside extracellular vesicles (EVs) for re-entry, while direct cell-cell contact is hardly involved. EVs account for a significant fraction of NP intercellular exchange, and its importance in NP transport is demonstrated in vitro and in vivo. While freely released NPs engage with the same receptors for re-entry, EV-enclosed ones bypass this dependence. These studies provide an easy and precise system to investigate the intercellular exchange stage of NP delivery, and shed the first light in the importance of EVs in NP transport between cells and into complex tissues.
Collapse
Affiliation(s)
- Xian Wu
- Department of PharmaceuticsUniversity of MinnesotaMinneapolisMN55455USA
| | - Tang Tang
- Department of PharmaceuticsUniversity of MinnesotaMinneapolisMN55455USA
| | - Yushuang Wei
- Department of PharmaceuticsUniversity of MinnesotaMinneapolisMN55455USA
| | - Katherine A. Cummins
- Department of Biomedical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
| | - David K. Wood
- Department of Biomedical EngineeringUniversity of MinnesotaMinneapolisMN55455USA
| | - Hong‐Bo Pang
- Department of PharmaceuticsUniversity of MinnesotaMinneapolisMN55455USA
| |
Collapse
|
3
|
Pang L, Huang X, Zhu L, Xiao H, Li M, Guan H, Gao J, Jin H. [Targeted killing of CD133 + lung cancer stem cells using paclitaxel-loaded PLGA-PEG nanoparticles with CD133 aptamers]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:26-35. [PMID: 35249867 DOI: 10.12122/j.issn.1673-4254.2022.01.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To construct a polylactic acid-glycolic acid-polyethylene glycol (PLGA-PEG) nanocarrier (N-Pac-CD133) coupled with a CD133 nucleic acid aptamer carrying paclitaxel for eliminating lung cancer stem cells (CSCs). METHODS Paclitaxel-loaded N-Pac-CD133 was prepared using the emulsion/solvent evaporation method and characterized. CD133+ lung CSCs were separated by magnetic bead separation and identified for their biological behaviors and gene expression profile. The efficiency of paclitaxel-loaded N-Pac-CD133 for targeted killing of lung cancer cells was assessed in vitro. SCID mice were inoculated with A549 cells and received injections of normal saline, empty nanocarrier linked with CD133 aptamer (N-CD133), paclitaxel, paclitaxel-loaded nanocarrier (N-Pac) or paclitaxel-loaded N-Pac-CD133 (n=8, 5 mg/kg paclitaxel) on days 10, 15 and 20, and the tumor weight and body weight of the mice were measured on day 40. RESULTS Paclitaxel-loaded N-Pac-CD133 showed a particle size of about 100 nm with a high encapsulation efficiency (>80%) and drug loading rate (>8%), and was capable of sustained drug release within 48 h. The CD133+ cell population in lung cancer cells showed the characteristic features of lung CSCs, including faster growth rate (30 days, P=0.001) and high expressions of tumor stem cell markers OV6(P < 0.001), CD133 (P=0.001), OCT3/4 (P=0.002), EpCAM (P=0.04), NANOG (P=0.005) and CD44 (P=0.02). Compared with N-Pac and free paclitaxel, paclitaxel-loaded N-Pac-CD133 showed significantly enhanced targeting ability and cytotoxicity against lung CSCs in vitro (P < 0.001) and significantly reduced the formation of tumor spheres (P < 0.001). In the tumor-bearing mice, paclitaxel-loaded N-Pac-CD133 showed the strongest effects in reducing the tumor mass among all the treatments (P < 0.001). CONCLUSION CD133 aptamer can promote targeted delivery of paclitaxel to allow targeted killing of CD133+ lung CSCs. N-Pac-CD133 loaded with paclitaxel may provide an effective treatment for lung cancer by targeting the lung cancer stem cells.
Collapse
Affiliation(s)
- L Pang
- First School of Clinical Medicine, Mudanjiang Medical University, Mudanjiang 157011, China
| | - X Huang
- Department of Respiratory and Critical Care Medicine, Wuhan First Hospital, Wuhan 430022, China
| | - L Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang 157011, China
| | - H Xiao
- Research Department, Mudanjiang Medical University, Mudanjiang 157011, China
| | - M Li
- First School of Clinical Medicine, Mudanjiang Medical University, Mudanjiang 157011, China
| | | | - J Gao
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - H Jin
- Clinical Laboratory, 5Department of Hematology, Affiliated Hongqi Hospital, Mudanjiang Medical University, Mudanjiang 157011, China
| |
Collapse
|
4
|
Cancer Stem Cells and Their Vesicles, Together with Other Stem and Non-Stem Cells, Govern Critical Cancer Processes: Perspectives for Medical Development. Int J Mol Sci 2022; 23:ijms23020625. [PMID: 35054811 PMCID: PMC8775347 DOI: 10.3390/ijms23020625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/17/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
Stem cells, identified several decades ago, started to attract interest at the end of the nineties when families of mesenchymal stem cells (MSCs), concentrated in the stroma of most organs, were found to participate in the therapy of many diseases. In cancer, however, stem cells of high importance are specific to another family, the cancer stem cells (CSCs). This comprehensive review is focused on the role and the mechanisms of CSCs and of their specific extracellular vesicles (EVs), which are composed of both exosomes and ectosomes. Compared to non-stem (normal) cancer cells, CSCs exist in small populations that are preferentially distributed to the niches, such as minor specific tissue sites corresponding to the stroma of non-cancer tissues. At niches and marginal sites of other cancer masses, the tissue exhibits peculiar properties that are typical of the tumor microenvironment (TME) of cancers. The extracellular matrix (ECM) includes components different from non-cancer tissues. CSCs and their EVs, in addition to effects analogous to those of MSCs/EVs, participate in processes of key importance, specific to cancer: generation of distinct cell subtypes, proliferation, differentiation, progression, formation of metastases, immune and therapy resistance, cancer relapse. Many of these, and other, effects require CSC cooperation with surrounding cells, especially MSCs. Filtered non-cancer cells, especially macrophages and fibroblasts, contribute to collaborative cancer transition/integration processes. Therapy developments are mentioned as ongoing preclinical initiatives. The preliminary state of clinical medicine is presented in terms of both industrial development and future treatments. The latter will be administered to specific patients together with known drugs, with the aim of eradicating their tumor growth and metastases.
Collapse
|
5
|
Effects of D-α-tocopherol polyethylene glycol succinate-emulsified poly(lactic-co-glycolic acid) nanoparticles on the absorption, pharmacokinetics, and pharmacodynamics of salinomycin sodium. Anticancer Drugs 2020; 30:72-80. [PMID: 30239423 DOI: 10.1097/cad.0000000000000695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although salinomycin sodium (SS) has shown in-vitro potential to inhibit cancer stem cell growth and development, its low water solubility makes it a poor candidate as an oral chemotherapeutic agent. To improve the bioavailability of SS, SS was encapsulated here using D-α-tocopherol polyethylene glycol succinate (TPGS)-emulsified poly(lactic-co-glycolic acid) (PLGA) nanoparticles and compared with its parent SS in terms of absorption, pharmacokinetics, and efficacy in suppressing nasopharyngeal carcinomas stem cells. The pharmacokinetics of SS and salinomycin sodium-loaded D-α-tocopherol polyethylene glycol succinate-emulsified poly(lactic-co-glycolic acid) nanoparticles (SLN) prepared by nanoprecipitation were analyzed in-vivo by timed-interval blood sampling and oral administration of SS and SLN to rats. Sensitive liquid chromatography-mass spectrometry (LC-MS) was developed to quantify plasma drug concentrations. SS and SLN transport in Caco-2 cells was also investigated. The therapeutic efficacy of SS and SLN against cancer stem cells was determined by orally administering the drugs to mice bearing CNE1 and CNE2 nasopharyngeal carcinoma xenografts and then evaluating CD133 cell proportions and tumorsphere formation. The in-vivo trial with rats showed that the Cmax, AUC(0-t), and Tmax for orally administered SLN were all significantly higher than those for SS (P<0.05). These findings were corroborated by a Caco-2 cell Transwell assay showing that relative SLN absorption was greater than that of SS on the basis of their apparent permeability coefficients (Papp). Significantly, therapeutic SLN efficacy against nasopharyngeal carcinoma stem cells was superior to that of SS. TPGS-emulsified PLGA nanoparticles effectively increase SS solubility and bioavailability. SLN is, therefore, promising as an oral chemotherapeutic agent against cancer stem cells.
Collapse
|
6
|
Wang Q, Liu F, Wang L, Xie C, Wu P, Du S, Zhou S, Sun Z, Liu Q, Yu L, Liu B, Li R. Enhanced and Prolonged Antitumor Effect of Salinomycin-Loaded Gelatinase-Responsive Nanoparticles via Targeted Drug Delivery and Inhibition of Cervical Cancer Stem Cells. Int J Nanomedicine 2020; 15:1283-1295. [PMID: 32161458 PMCID: PMC7049776 DOI: 10.2147/ijn.s234679] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/26/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cervical cancer stem cells (CCSCs) represent a subpopulation of tumor cells that possess self-renewal capacity and numerous intrinsic mechanisms of resistance to conventional chemotherapy and radiotherapy. These cells play a crucial role in relapse and metastasis of cervical cancer. Therefore, eradication of CCSCs is the primary objective in cervical cancer therapy. Salinomycin (Sal) is an agent used for the elimination of cancer stem cells (CSCs); however, the occurrence of several side effects hinders its application. Nanoscale drug-delivery systems offer great promise for the diagnosis and treatment of tumors. These systems can be used to reduce the side effects of Sal and improve clinical benefit. METHODS Sal-loaded polyethylene glycol-peptide-polycaprolactone nanoparticles (Sal NPs) were fabricated under mild and non-toxic conditions. The real-time biodistribution of Sal NPs was investigated through non-invasive near-infrared fluorescent imaging. The efficacy of tumor growth inhibition by Sal NPs was evaluated using tumor xenografts in nude mice. Flow cytometry, immunohistochemistry, and Western blotting were used to detect the apoptosis of CSCs after treatment with Sal NPs. Immunohistochemistry and Western blotting were used to examine epithelial-mesenchymal transition (epithelial interstitial transformation) signal-related molecules. RESULTS Sal NPs exhibited antitumor efficacy against cervical cancers by inducing apoptosis of CCSCs and inhibiting the epithelial-mesenchymal transition pathway. Besides, tumor pieces resected from Sal NP-treated mice showed decreased reseeding ability and growth speed, further demonstrating the significant inhibitory ability of Sal NPs against CSCs. Moreover, owing to targeted delivery based on the gelatinase-responsive strategy, Sal NPs was more effective and tolerable than free Sal. CONCLUSION To the best of our knowledge, this is the first study to show that CCSC-targeted Sal NPs provide a potential approach to selectively target and efficiently eradicate CCSCs. This renders them a promising strategy to improve the therapeutic effect against cervical cancer.
Collapse
Affiliation(s)
- Qin Wang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Fangcen Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Lifeng Wang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Chen Xie
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing210023, People’s Republic of China
| | - Puyuan Wu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Shiyao Du
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Shujuan Zhou
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Zhichen Sun
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Qin Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Rutian Li
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| |
Collapse
|
7
|
Xu J, Jing M, Wang S, Yang C, Chen X. A review of medical image detection for cancers in digestive system based on artificial intelligence. Expert Rev Med Devices 2019; 16:877-889. [PMID: 31530047 DOI: 10.1080/17434440.2019.1669447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: At present, cancer imaging examination relies mainly on manual reading of doctors, which requests a high standard of doctors' professional skills, clinical experience, and concentration. However, the increasing amount of medical imaging data has brought more and more challenges to radiologists. The detection of digestive system cancer (DSC) based on artificial intelligence (AI) can provide a solution for automatic analysis of medical images and assist doctors to achieve high-precision intelligent diagnosis of cancers. Areas covered: The main goal of this paper is to introduce the main research methods of the AI based detection of DSC, and provide relevant reference for researchers. Meantime, it summarizes the main problems existing in these methods, and provides better guidance for future research. Expert commentary: The automatic classification, recognition, and segmentation of DSC can be better realized through the methods of machine learning and deep learning, which minimize the internal information of images that are difficult for humans to discover. In the diagnosis of DSC, the use of AI to assist imaging surgeons can achieve cancer detection rapidly and effectively and save doctors' diagnosis time. These can lay the foundation for better clinical diagnosis, treatment planning and accurate quantitative evaluation of DSC.
Collapse
Affiliation(s)
- Jiangchang Xu
- Institute of Biomedical Manufacturing and Life Quality Engineering, State Key Laboratory of Mechanical System and Vibration, School of Mechanical Engineering, Shanghai Jiao Tong University , Shanghai , China
| | - Mengjie Jing
- Institute of Biomedical Manufacturing and Life Quality Engineering, State Key Laboratory of Mechanical System and Vibration, School of Mechanical Engineering, Shanghai Jiao Tong University , Shanghai , China
| | - Shiming Wang
- Institute of Biomedical Manufacturing and Life Quality Engineering, State Key Laboratory of Mechanical System and Vibration, School of Mechanical Engineering, Shanghai Jiao Tong University , Shanghai , China
| | - Cuiping Yang
- Department of Gastroenterology, Ruijin North Hospital of Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Xiaojun Chen
- Institute of Biomedical Manufacturing and Life Quality Engineering, State Key Laboratory of Mechanical System and Vibration, School of Mechanical Engineering, Shanghai Jiao Tong University , Shanghai , China
| |
Collapse
|
8
|
Chen D, Pan X, Xie F, Lu Y, Zou H, Yin C, Zhang Y, Gao J. Codelivery of doxorubicin and elacridar to target both liver cancer cells and stem cells by polylactide-co-glycolide/d-alpha-tocopherol polyethylene glycol 1000 succinate nanoparticles. Int J Nanomedicine 2018; 13:6855-6870. [PMID: 30498347 PMCID: PMC6207383 DOI: 10.2147/ijn.s181928] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Purpose Liver cancer is the third leading cause of cancer-related deaths worldwide. Liver cancer stem cells (LCSCs) are a subpopulation of cancer cells that are responsible for the initiation, progression, drug resistance, recurrence, and metastasis of liver cancer. Recent studies have suggested that the eradication of both LCSCs and liver cancer cells is necessary because the conversion of cancer stem cells (CSCs) to cancer cells occasionally occurs. As ATP-binding cassette (ABC) transporters are overexpressed in both CSCs and cancer cells, combined therapies using ABC transporter inhibitors and chemotherapy drugs could show superior therapeutic efficacy in liver cancer. In this study, we developed poly(lactide-co-glycolide)/d-alpha-tocopherol polyethylene glycol 1000 succinate nanoparticles to accomplish the simultaneous delivery of an optimized ratio of doxorubicin (DOX) and elacridar (ELC) to target both LCSCs and liver cancer cells. Methods Median-effect analysis was used for screening of DOX and ELC for synergy in liver cancer cells (HepG2 cells) and LCSCs (HepG2 tumor sphere [HepG2-TS]). Then, nanoparticles loaded with DOX and ELC at the optimized ratio (NDEs) were prepared by nanoprecipitation method. The cytotoxicity and colony and tumor sphere formation ability of nanoparticles were investigated in vitro, and the tissue distribution and antitumor activity of nanoparticles were evaluated in vivo. Results We demonstrated that a DOX/ELC molar ratio of 1:1 was synergistic in HepG2 cells and HepG2-TS. NDEs were shown to exhibit significantly increased cytotoxic effects against both HepG2 and HepG2-TS compared with DOX-loaded nanoparticles (NDs) or ELC-loaded nanoparticles (NEs) in vitro. In vivo studies demonstrated that the nanoparticles exhibited better tumor targeting, with NDE showing the strongest antitumor activity with lower systemic toxicity. Conclusion These results suggested that NDE represented a promising combination therapy against liver cancer by targeting both liver cancer cells and CSCs.
Collapse
Affiliation(s)
- Dazhong Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China, .,Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China, .,Department of Planning, Kunming General Hospital of Chengdu Military Command, Yunnan 650032, China
| | - Xiaoli Pan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fangyuan Xie
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China
| | - Ying Lu
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China,
| | - Hao Zou
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China,
| | - Chuan Yin
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yu Zhang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei 441000, China,
| | - Jie Gao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China, .,Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China,
| |
Collapse
|
9
|
Sun N, Jiang T, Wu C, Sun H, Zhou Q, Lu L. Expression and influence of BMP-4 in human dental pulp cells cultured in vitro. Exp Ther Med 2018; 16:5112-5116. [PMID: 30542466 PMCID: PMC6257597 DOI: 10.3892/etm.2018.6824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 08/29/2018] [Indexed: 12/31/2022] Open
Abstract
Effects of bone morphogenetic protein (BMP)-4 on proliferation and differentiation capacities of dental pulp cells through BMP-4 acting on human dental pulp cells cultured in vitro were investigated. Dental pulp tissues of lesion-free teeth extracted from patients due to orthodontics were taken, and human dental pulp cells were cultured in vitro using the tissue explant method. Immunocytochemical staining was used for the identification of vimentin and keratin. The dental pulp cells were divided into groups A and B. A total of 100 ng/ml BMP-4 was added into group A, while no inducer was added into group B as the control group. The cell growth curves at day 1, 2, 3, 5 and 7 after culture were drawn. At day 7, the cell count, alkaline phosphatase (ALP) activity, number of calcified nodules, and expression levels of dentin sialophosphoprotein (DSPP), dentin matrix protein-1 (DMP-1) and each gene related to dentinogenesis in each group were detected, respectively. Human dental pulp cells were conformed to the biological characteristics of dental pulp cells according to the identification of vimentin and keratin via immunocytochemical staining. With the prolongation of culture time, the number of cells in both groups was gradually increased, reaching the peak at day 5 and began to decline at day 7. The number of cells in group A was significantly greater than that in group B (p<0.05). According to the results of reverse transcription-quantitative polymerase chain reaction (RT-qPCR), the relative messenger ribonucleic acid (mRNA) expression levels of ALP, DSPP and DMP-1 in group A were significantly higher than those in group B (p<0.05). BMP-4 can promote the growth of dental pulp cells and remarkably enhance the differentiation of dental pulp cells into odontoblasts.
Collapse
Affiliation(s)
- Ningning Sun
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Tianjiao Jiang
- Department of Oral Medicine, The Second Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Chuanbin Wu
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Haijiang Sun
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Qing Zhou
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Li Lu
- Department of Oral and Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| |
Collapse
|
10
|
Chen X, Zhang Z, Yang S, Chen H, Wang D, Li J. All-trans retinoic acid-encapsulated, CD20 antibody-conjugated poly(lactic- co-glycolic acid) nanoparticles effectively target and eliminate melanoma-initiating cells in vitro. Onco Targets Ther 2018; 11:6177-6187. [PMID: 30288053 PMCID: PMC6163018 DOI: 10.2147/ott.s169957] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose Melanoma, which is initiated from melanocytes, is the most fatal type of skin cancer. Melanoma-initiating cells significantly contribute to the initiation, metastasis, and recurrence of melanoma, and CD20 is a marker of melanoma-initiating cells. All-trans retinoic acid (ATRA) has been demonstrated to induce differentiation, inhibit proliferation, and promote the apoptosis of cancer cells and cancer-initiating cells (CICs). However, there has been no report on ATRA activity against melanoma-initiating cells. In this study, we examined the activity of ATRA against melanoma-initiating cells and developed ATRA-encapsulated poly(lactic-co-glycolic acid) (PLGA) nanoparticles, which were conjugated with a CD20 antibody (ATRA-PNP-CD20) for targeted delivery of ATRA to CD20+ melanoma-initiating cells. Materials and methods The effects of ATRA and ATRA-PNP-CD20 against melanoma-initiating cells were investigated using a cytotoxicity assay, tumorsphere formation assay, and flow cytometry. Results ATRA-PNP-CD20 had a size of 126.9 nm and a negative zeta potential. The drug-loading capacity of ATRA-PNP-CD20 was 8.7%, and ATRA-PNP-CD20 displayed a sustained release of ATRA for 144 hours. The results showed that ATRA-PNP-CD20 could effectively and specifically deliver ATRA to CD20+ melanoma-initiating cells, achieving superior inhibitory effects against CD20+ melanoma-initiating cells compared with those of free ATRA and nontargeted nanoparticles. To the best of our knowledge, we report for the first time a potent activity of ATRA against CD20+ melanoma-initiating cells, targeted drug delivery of ATRA via nanoparticles to melanoma-initiating cells, and the achievement of a superior inhibitory effect against melanoma-initiating cells by using a CD20 antibody. Conclusion ATRA-PNP-CD20 represents a promising tool for eliminating melanoma-initiating cells and shows a potential for the therapy of melanoma.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Dermatology and Venerology, Shandong University School of Medicine, Jinan, Shandong, 250000, China, .,Department of Dermatology, Qingdao Municipal Hospital, Qingdao, Shandong 266011, China,
| | - Zhiyuan Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Shengfeng Yang
- Department of Medical Oncology, Qingdao Center Hospital, Qingdao, Shandong 266011, China
| | - Hairong Chen
- Department of Dermatology, Qingdao Municipal Hospital, Qingdao, Shandong 266011, China,
| | - Dan Wang
- Department of Ultrasound, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Jun Li
- College of Pharmacy, Liaocheng University, Liaocheng, Shandong 252000, China,
| |
Collapse
|
11
|
Zhang Y, Zhao J, Sun J, Huang L, Li Q. Targeting lung cancer initiating cells by all-trans retinoic acid-loaded lipid-PLGA nanoparticles with CD133 aptamers. Exp Ther Med 2018; 16:4639-4649. [PMID: 30542415 PMCID: PMC6257334 DOI: 10.3892/etm.2018.6762] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
Lung cancer initiating cells represent a specific subpopulation of lung cancer cells, which significantly contribute to the initiation, metastasis and recurrence of lung cancer. CD133, initially considered a marker of stem cells, is now considered as a marker for lung cancer initiating cells. All-trans retinoic acid (RA) has been demonstrated to cause the differentiation, inhibition of proliferation, and apoptosis of cancer cells and cancer initiating cells. However, there have been no reports on the activity of RA against lung cancer initiating cells. In the present study, the activity of RA against lung cancer initiating cells was investigated by determining the cytotoxicity, and performing a tumorsphere assay and flow cytometry-based analysis. In addition, to promote the therapeutic effect of RA in CD133+ lung cancer initiating cells, RA-loaded lipid poly(lactic-co-glycolic acid) (PLGA) nanoparticles with CD133 aptamers (RA-LPNPs-CD133) were developed. The activity of RA and RA-LPNPs-CD133 against lung cancer initiating cells was also investigated. RA-LPNPs-CD133 had a size of 129.9 nm, and exhibited sustained release of RA during the 144-h period. For the first time, to the best of our knowledge, the present study demonstrated that RA exerted potent activity towards CD133+ lung cancer initiating cells. The results also showed that RA-LPNPs-CD133 efficiently and specifically promoted the delivery of RA to CD133+ lung cancer initiating cells, exhibiting superior inhibitory effects against CD133+ lung cancer initiating cells compared with non-targeted nanoparticles and RA. To the best of our knowledge, the present study is the first to report the promotion of RA delivery via nanoparticles to lung cancer initiating cells and achievement of a superior inhibitory effect against lung cancer initiating cells by the utilization of CD133 aptamers. Therefore, RA-LPNPs-CD133 represents a promising tool for the elimination of lung cancer initiating cells.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Juan Zhao
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Jing Sun
- Department of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Lu Huang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| | - Qingfeng Li
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, P.R. China
| |
Collapse
|
12
|
Mi Y, Huang Y, Deng J. The enhanced delivery of salinomycin to CD133 + ovarian cancer stem cells through CD133 antibody conjugation with poly(lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles. Oncol Lett 2018; 15:6611-6621. [PMID: 29725407 DOI: 10.3892/ol.2018.8140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 11/20/2017] [Indexed: 01/02/2023] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy, and ovarian cancer stem cells (CSCs) serve a pivotal function in the metastasis and recurrence of ovarian cancer. Multiple previous studies have validated CD133 as a marker of ovarian CSCs. Although salinomycin is a promising therapeutic agent that has been demonstrated to kill CSCs in various types of cancer, poor aqueous solubility hampers its clinical application. The present study used salinomycin-loaded poly(lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles conjugated with CD133 antibodies (CD133-SAL-NP) to eliminate CD133+ ovarian CSCs. The results revealed that CD133-SAL-NPs were of an appropriate size (149.2 nm) and exhibited sustained drug release. CD133-SAL-NPs efficiently bound to CD133+ ovarian cancer cells, resulting in an increased cytotoxic effect in CD133+ ovarian cancer cells, compared with the untargeted SAL-NPs and salinomycin. CD133-SAL-NPs reduced the percentage of CD133+ ovarian CSCs in ovarian cells more effectively than treatment with salinomycin or SAL-NPs, suggesting that CD133-SAL-NP targeted CD133+ ovarian CSCs. In nude mice bearing ovarian cancer xenografts, CD133-SAL-NPs exerted improved therapeutic effects compared with SAL-NPs and salinomycin. Thus, CD133 was demonstrated to be a promising target for drug delivery to ovarian CSCs, and may be useful as an agent to inhibit the growth of ovarian cancer by targeting CD133+ ovarian CSCs. CD133-SAL-NPs may therefore represent a promising approach for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yi Mi
- Department of Obstetrics and Gynecology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| | - Yuqin Huang
- Department of Obstetrics and Gynecology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China.,Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jie Deng
- Department of Obstetrics and Gynecology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| |
Collapse
|
13
|
Salinomycin-loaded lipid-polymer nanoparticles with anti-CD20 aptamers selectively suppress human CD20+ melanoma stem cells. Acta Pharmacol Sin 2018; 39:261-274. [PMID: 29388568 DOI: 10.1038/aps.2017.166] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/18/2017] [Indexed: 12/15/2022]
Abstract
Melanoma is the deadliest type of skin cancer. CD20+ melanoma stem cells (CSCs) are pivotal for metastasis and initiation of melanoma. Therefore, selective elimination of CD20+ melanoma CSCs represents an effective treatment to eradicate melanoma. Salinomycin has emerged as an effective drug toward various CSCs. Due to its poor solubility, its therapeutic efficacy against melanoma CSCs has never been evaluated. In order to target CD20+ melanoma CSCs, we designed salinomycin-loaded lipid-polymer nanoparticles with anti-CD20 aptamers (CD20-SA-NPs). Using a single-step nanoprecipitation method, salinomycin-loaded lipid-polymer nanoparticles (SA-NPs) were prepared, then CD20-SA-NPs were obtained through conjugation of thiolated anti-CD20 aptamers to SA-NPs via a maleimide-thiol reaction. CD20-SA-NPs displayed a small size of 96.3 nm, encapsulation efficiency higher than 60% and sustained drug release ability. The uptake of CD20-SA-NPs by CD20+ melanoma CSCs was significantly higher than that of SA-NPs and salinomycin, leading to greatly enhanced cytotoxic effects in vitro, thus the IC50 values of CD20-SA-NPs were reduced to 5.7 and 2.6 μg/mL in A375 CD+20 cells and WM266-4 CD+ cells, respectively. CD20-SA-NPs showed a selective cytotoxicity toward CD20+ melanoma CSCs, as evidenced by the best therapeutic efficacy in suppressing the formation of tumor spheres and the proportion of CD20+ cells in melanoma cell lines. In mice bearing melanoma xenografts, administration of CD20-SA-NPs (salinomycin 5 mg·kg-1·d-1, iv, for 60 d) showed a superior efficacy in inhibition of melanoma growth compared with SA-NPs and salinomycin. In conclusion, CD20 is a superior target for delivering drugs to melanoma CSCs. CD20-SA-NPs display effective delivery of salinomycin to CD20+ melanoma CSCs and represent a promising treatment for melanoma.
Collapse
|
14
|
Zhang Y, Zhang Q, Sun J, Liu H, Li Q. The combination therapy of salinomycin and gefitinib using poly(d,l-lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles for targeting both lung cancer stem cells and cancer cells. Onco Targets Ther 2017; 10:5653-5666. [PMID: 29225473 PMCID: PMC5709995 DOI: 10.2147/ott.s141083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Purpose Lung cancer (LC) is the leading cause of cancer death worldwide. Evidences suggest that both LC cancer stem cells (CSCs) and cancer cells are supposed to be eliminated to achieve superior treatment effect against LC. Salinomycin could eradiate CSCs in various types of cancers, and gefitinib is a first-line therapy in LC. The purpose of the present study was to develop salinomycin-loaded nanoparticles (salinomycin-NPs) combined with gefitinib-loaded nanoparticles (gefitinib-NPs) to eradicate both LC CSCs and cancer cells. Methods Salinomycin and gefitinib were encapsulated separately by poly(d,l-lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles by the emulsion/solvent evaporation approach. The anti-LC activity of salinomycin-NPs and gefitinib-NPs was investigated. Results Salinomycin-NPs and gefitinib-NPs are of ~140 nm in size, high drug encapsulation efficacy and sustained release of drugs. CD133+ LC CSCs showed the characteristics of CSCs, including significantly enhanced stem cell gene expression, tumorsphere formation ability, and tumorigenicity in mice. Both salinomycin and salinomycin-NPs are capable of selectively inhibiting LC CSCs, as reflected by their enhanced cytotoxic effects toward CD133+ LC CSCs and ability to reduce tumorsphere formation in LC cell lines, whereas gefitinib and gefitinib-NPs could significantly inhibit LC cells. Salinomycin-NPs and salinomycin could reduce the population of LC CSCs in the tumors in vivo. It is noteworthy that salinomycin-NPs combined with gefitinib-NPs inhibited the growth of tumors more efficiently compared with salinomycin combined with gefitinib or single salinomycin-NPs or gefitinib-NPs. Conclusion Salinomycin-NPs combined with gefitinib-NPs represent a potential approach for LC by inhibiting both LC CSCs and cancer cells.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Oncology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei College of Arts and Science, Xiangyang 441000, Hubei, China
| | - Qi Zhang
- Department of Oncology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei College of Arts and Science, Xiangyang 441000, Hubei, China
| | - Jing Sun
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Huijie Liu
- Department of Oncology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei College of Arts and Science, Xiangyang 441000, Hubei, China
| | - Qingfeng Li
- Department of Oncology, Xiangyang Central Hospital, The Affiliated Hospital of Hubei College of Arts and Science, Xiangyang 441000, Hubei, China
| |
Collapse
|
15
|
Ma J, Zhuang H, Zhuang Z, Lu Y, Xia R, Gan L, Wu Y. Development of docetaxel liposome surface modified with CD133 aptamers for lung cancer targeting. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1864-1871. [PMID: 29082764 DOI: 10.1080/21691401.2017.1394874] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Junzhe Ma
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Huiru Zhuang
- Department of Plastic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Yufeng Lu
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Rui Xia
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Lei Gan
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Yuantao Wu
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| |
Collapse
|
16
|
Codelivery of salinomycin and docetaxel using poly(D,L-lactic-co-glycolic acid)-poly(ethylene glycol) nanoparticles to target both gastric cancer cells and cancer stem cells. Anticancer Drugs 2017; 28:989-1001. [DOI: 10.1097/cad.0000000000000541] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
17
|
Guo X, Zhu X, Gao J, Liu D, Dong C, Jin X. PLGA nanoparticles with CD133 aptamers for targeted delivery and sustained release of propranolol to hemangioma. Nanomedicine (Lond) 2017; 12:2611-2624. [PMID: 28960167 DOI: 10.2217/nnm-2017-0130] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM To develop propranolol-loaded poly(lactic-co-glycolic acid) nanoparticle with CD133 aptamers (PPN-CD133) to treat infantile hemangioma. MATERIALS & METHODS The antihemangioma activity and mechanism of PPN-CD133 were evaluated. RESULTS & CONCLUSION PPN-CD133 are of desired size (143.7 nm), drug encapsulation efficiency (51.8%) and sustained drug release for 8 days. PPN-CD133 could effectively bind to CD133+ hemangioma stem cells, resulting in enhanced cytotoxic effect and reduced expression of angiogenesis factors in hemangioma stem cells. The therapeutic effect of PPN-CD133 in hemangioma was superior to that of untargeted PPN and propranolol in vivo, as reflected by reduced hemangioma volume, weight and microvessel density. PPN-CD133 represents a very promising approach to locally and efficiently deliver propranolol leading to significant inhibition of infantile hemangioma.
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwuweiqi Road, Ji'nan 250021, China
| | - Xiaoshuang Zhu
- Department of Hemangioma & Vascular Malformation, He'nan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China
| | - Jie Gao
- Department of Pharmacy Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Dakan Liu
- Department of Hemangioma & Vascular Malformation, He'nan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China
| | - Changxian Dong
- Department of Hemangioma & Vascular Malformation, He'nan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou 450003, China
| | - Xing Jin
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, 324 Jingwuweiqi Road, Ji'nan 250021, China
| |
Collapse
|
18
|
Guo X, Zhu X, Liu D, Gong Y, Sun J, Dong C. Continuous delivery of propranolol from liposomes-in-microspheres significantly inhibits infantile hemangioma growth. Int J Nanomedicine 2017; 12:6923-6936. [PMID: 29075111 PMCID: PMC5609781 DOI: 10.2147/ijn.s137634] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose To reduce the adverse effects and high frequency of administration of propranolol to treat infantile hemangioma, we first utilized propranolol-loaded liposomes-in-microsphere (PLIM) as a novel topical release system to realize sustained release of propranolol. Methods PLIM was developed from encapsulating propranolol-loaded liposomes (PLs) in microspheres made of poly(lactic-co-glycolic acid)-b-poly(ethylene glycol)-b-poly(lactic-co-glycolic acid) copolymers (PLGA-PEG-PLGA). The release profile of propranolol from PLIM was evaluated, and its biological activity was investigated in vitro using proliferation assays on hemangioma stem cells (HemSCs). Tumor inhibition was studied in nude mice bearing human subcutaneous infantile hemangioma. Results The microspheres were of desired particle size (~77.8 μm) and drug encapsulation efficiency (~23.9%) and achieved sustained drug release for 40 days. PLIM exerted efficient inhibition of the proliferation of HemSCs and significantly reduced the expression of two angiogenesis factors (vascular endothelial growth factor-A [VEGF-A] and basic fibroblast growth factor [bFGF]) in HemSCs. Notably, the therapeutic effect of PLIM in hemangioma was superior to that of propranolol and PL in vivo, as reflected by significantly reduced hemangioma volume, weight, and microvessel density. The mean hemangioma weight of the PLIM-treated group was significantly lower than that of other groups (saline =0.28 g, propranolol =0.21 g, PL =0.13 g, PLIM =0.03 g; PLIM vs saline: P<0.001, PLIM vs propranolol: P<0.001, PLIM vs PL: P<0.001). The mean microvessel density of the PLIM-treated group was significantly lower than that of other groups (saline =40 vessels/mm2, propranolol =31 vessels/mm2, PL =25 vessels/mm2, PLIM =11 vessels/mm2; PLIM vs saline: P<0.001, PLIM vs propranolol: P<0.01, PLIM vs PL: P<0.05). Conclusion Our findings show that PLIM is a very promising approach to locally and efficiently deliver propranolol to the hemangioma site leading to a significant inhibition of infantile hemangioma.
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Xiaoshuang Zhu
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Dakan Liu
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Yubin Gong
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Jing Sun
- Department of Pharmacy, Second Military Medical University, Shanghai, People's Republic of China
| | - Changxian Dong
- Department of Hemangioma and Vascular Malformation, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| |
Collapse
|
19
|
Huang X, Huang J, Leng D, Yang S, Yao Q, Sun J, Hu J. Gefitinib-loaded DSPE-PEG2000 nanomicelles with CD133 aptamers target lung cancer stem cells. World J Surg Oncol 2017; 15:167. [PMID: 28854941 PMCID: PMC5577827 DOI: 10.1186/s12957-017-1230-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/13/2017] [Indexed: 11/25/2022] Open
Abstract
Background Lung cancer stem cells (CSCs) are considered to be the seed of lung cancer, and CD133 is a marker of lung CSCs. Here, we developed gefitinib-loaded poly(ethylene glycol) 2000-distearoylphosphatidylethanolamine nanomicelles with CD133 aptamers (M-Gef-CD133) to eliminate CD133+ lung CSCs. Methods M-Gef-CD133 was prepared using a lipid-film-based approach. The targeting and activity of M-Gef-CD133 towards lung CSCs were evaluated. Results M-Gef-CD133 were small (25 nm) and showed enhanced cytotoxic effect towards CD133+ lung CSCs compared with non-targeted M-Gef and gefitinib. Notably, M-Gef-CD133 could significantly reduce tumor sphere formation and the percentage of CD133+ lung CSCs, indicating that it possesses selective toxicity against CD133+ lung CSCs. Conclusions The interaction of CD133 aptamers and CD133 shows promise in the delivery of gefitinib to CD133+ lung CSCs, and M-Gef-CD133 represents a promising treatment to target lung CSCs.
Collapse
Affiliation(s)
- Xiaolong Huang
- Department of Respiratory Medicine, Wuhan NO. 1 Hospital, 215 Zhongshan Street, Wuhan, 430022, China.
| | - Jingsong Huang
- Laboratory Medicine, Third Hubei Provincial People's Hospital, Zhongshan Street, Wuhan, 430022, China
| | - Dewen Leng
- Department of Critical Care Medicine, Wuhan NO. 1 Hospital, 215 Zhongshan Street, Wuhan, 430022, China
| | - Shuo Yang
- Department of Respiratory Medicine, Wuhan NO. 1 Hospital, 215 Zhongshan Street, Wuhan, 430022, China
| | - Qi Yao
- Department of Otolaryngology, Wuhan NO. 1 Hospital, 215 Zhongshan Street, Wuhan, 430022, China
| | - Jin Sun
- Department of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Jun Hu
- Department of Physical examination, Wuhan Hospital for Occupational Disease Prevention and Treatment, 18-20 Jianghanbei Road, Wuhan, 430016, China
| |
Collapse
|
20
|
Wang M, Xie F, Wen X, Chen H, Zhang H, Liu J, Zhang H, Zou H, Yu Y, Chen Y, Sun Z, Wang X, Zhang G, Yin C, Sun D, Gao J, Jiang B, Zhong Y, Lu Y. Therapeutic PEG-ceramide nanomicelles synergize with salinomycin to target both liver cancer cells and cancer stem cells. Nanomedicine (Lond) 2017; 12:1025-1042. [PMID: 28440698 DOI: 10.2217/nnm-2016-0408] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM Salinomycin (SAL)-loaded PEG-ceramide nanomicelles (SCM) were prepared to target both liver cancer cells and cancer stem cells. MATERIALS & METHODS The synergistic ratio of SAL/PEG-ceramide was evaluated to prepare SCM, and the antitumor activity of SCM was examined both in vitro and in vivo. RESULTS SAL/PEG-ceramide molar ratio of 1:4 was chosen as the synergistic ratio, and SCM showed superior cytotoxic effect and increased apoptosis-inducing activity in both liver cancer cells and cancer stem cells. In vivo, SCM showed the best tumor inhibitory effect with a safety profile. CONCLUSION Thus, PEG-ceramide nanomicelles could serve as an effective and safe therapeutic drug carrier to deliver SAL into liver cancer, opening up the avenue of using PEG-ceramide as therapeutic drug carriers.
Collapse
Affiliation(s)
- Meiping Wang
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Fangyuan Xie
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Xikai Wen
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Han Chen
- Department of General Surgery, 411 Hospital of Chinese People's Liberation Army, 15 East Jiangwan Road, Shanghai 200081, China
| | - Hai Zhang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Junjie Liu
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - He Zhang
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Hao Zou
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yuan Yu
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yan Chen
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Zhiguo Sun
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Xinxia Wang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Guoqing Zhang
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, 225 Changhai Road, Shanghai 200438, China
| | - Chuan Yin
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Duxin Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jie Gao
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Beige Jiang
- Third Department of HepaticSurgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China
| | - Yanqiang Zhong
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Ying Lu
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
21
|
Zhu M, Chen S, Hua L, Zhang C, Chen M, Chen D, Dong Y, Zhang Y, Li M, Song X, Chen H, Zheng H. Self-targeted salinomycin-loaded DSPE-PEG-methotrexate nanomicelles for targeting both head and neck squamous cell carcinoma cancer cells and cancer stem cells. Nanomedicine (Lond) 2017; 12:295-315. [PMID: 28093940 DOI: 10.2217/nnm-2016-0382] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To target both head and neck squamous cell carcinoma (HNSCC) cells and cancer stem cells (CSCs) by salinomycin-loaded DSPE-PEG-MTX (synthesized using DSPE-PEG2000-NH2 and methotrexate) nanomicelles (M-SAL-MTX). MATERIALS & METHODS The characterization, antitumor activity and mechanism of M-SAL-MTX were evaluated. RESULTS & CONCLUSION M-SAL-MTX showed enhanced inhibitory effect toward both HNSCC CSCs and non-CSCs compared with a single treatment of methotrexate and salinomycin. In nude mice-bearing HNSCC xenografts, M-SAL-MTX suppressed tumor growth more effectively than other controls including combination of methotrexate and salinomycin. Therefore, M-SAL-MTX may provide a strategy for treating HNSCC by targeting both HNSCC CSCs and HNSCC cells.
Collapse
Affiliation(s)
- Minhui Zhu
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Shicai Chen
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Libo Hua
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Caiyun Zhang
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Mengjie Chen
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Donghui Chen
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yinmei Dong
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Yingying Zhang
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Meng Li
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Xianmin Song
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Huaiwen Chen
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China.,Sunlipo Biotech Research Center for Nanomedicine, 3688 Tingwei Road, Shanghai 201507, China
| | - Hongliang Zheng
- Department of Otolaryngology Head & Neck Surgery, Shanghai Changhai Hospital, the Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|