1
|
Shi H, Wang R, Cao HC, Guo HY, Pan P, Xiong CF, Zhang LJ, Yang Q, Wei S, Liu T. A Metal-Polyphenol-Based Oxygen Economizer and Fenton Reaction Amplifier for Self-Enhanced Synergistic Photothermal/Chemodynamic/Chemotherapy. Adv Healthc Mater 2023; 12:e2300054. [PMID: 36977362 DOI: 10.1002/adhm.202300054] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/11/2023] [Indexed: 03/30/2023]
Abstract
To overcome the limitations of doxorubicin (DOX) chemotherapy, nanomedicines that integrate additional photothermal therapy (PTT) and chemodynamic therapy (CDT) strategies are highlighted as promising alternatives for the treatment of malignant tumors. However, time-consuming preparation processes, biosafety concerns, and the bottlenecks of individual therapeutic modalities often limit the practical applications of this strategy. To address these issues, this work designs an oxygen economizer that additionally serves as a Fenton reaction amplifier through the simple assembly of epigallocatechin gallate (EGCG), pluronic F-127 (PF127), iron (III) ions, and doxorubicin (DOX) for the enhancement of synergistic PTT/CDT/chemotherapy. The resulting nanoformulation, EFPD, can target mitochondria and inhibit cell respiration to reduce O2 consumption, thus boosting DOX-mediated H2 O2 generation for enhanced CDT and simultaneously improving hypoxia-limited DOX chemotherapy efficacy. Moreover, the coordination between EGCG and Fe3+ provides EFPD with excellent photothermal conversion efficiencies (η = 34.7%) for PTT and photothermal-accelerated drug release. Experimental results indicate that EFPD-mediated synergistic enhancement of PTT/CDT/chemotherapy can achieve excellent therapeutic outcomes, including enhanced ablation of solid tumors, reduced metastasis and cardiotoxicity, and extended life spans.
Collapse
Affiliation(s)
- Hui Shi
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Ru Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Hu-Chen Cao
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Hai-Yan Guo
- School of Public Health, Anhui Medical University, Hefei, 230032, P. R. China
| | - Pei Pan
- School of Pharmacy, Anhui Medical University, Hefei, 230032, P. R. China
| | - Cheng-Feng Xiong
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Lin-Jun Zhang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Qiang Yang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Shuang Wei
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| | - Tao Liu
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, P. R. China
| |
Collapse
|
2
|
Yuan G, Liu Z, Wang W, Liu M, Xu Y, Hu W, Fan Y, Zhang X, Liu Y, Si G. Multifunctional nanoplatforms application in the transcatheter chemoembolization against hepatocellular carcinoma. J Nanobiotechnology 2023; 21:68. [PMID: 36849981 PMCID: PMC9969656 DOI: 10.1186/s12951-023-01820-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has the sixth-highest new incidence and fourth-highest mortality worldwide. Transarterial chemoembolization (TACE) is one of the primary treatment strategies for unresectable HCC. However, the therapeutic effect is still unsatisfactory due to the insufficient distribution of antineoplastic drugs in tumor tissues and the worsened post-embolization tumor microenvironment (TME, e.g., hypoxia and reduced pH). Recently, using nanomaterials as a drug delivery platform for TACE therapy of HCC has been a research hotspot. With the development of nanotechnology, multifunctional nanoplatforms have been developed to embolize the tumor vasculature, creating conditions for improving the distribution and bioavailability of drugs in tumor tissues. Currently, the researchers are focusing on functionalizing nanomaterials to achieve high drug loading efficacy, thorough vascular embolization, tumor targeting, controlled sustained release of drugs, and real-time imaging in the TACE process to facilitate precise embolization and enable therapeutic procedures follow-up imaging of tumor lesions. Herein, we summarized the recent advances and applications of functionalized nanomaterials based on TACE against HCC, believing that developing these functionalized nanoplatforms may be a promising approach for improving the TACE therapeutic effect of HCC.
Collapse
Affiliation(s)
- Gang Yuan
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China
| | - Zhiyin Liu
- grid.488387.8Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Weiming Wang
- grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China ,grid.488387.8Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Mengnan Liu
- grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China ,grid.488387.8National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yanneng Xu
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China
| | - Wei Hu
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China
| | - Yao Fan
- grid.410578.f0000 0001 1114 4286Department of Anus and Intestine Surgery, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China
| | - Xun Zhang
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China
| | - Yong Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Guangyan Si
- Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
Tao J, Wei Z, Cheng Y, Xu M, Li Q, Lee SMY, Ge W, Luo KQ, Wang X, Zheng Y. Apoptosis-Sensing Xenograft Zebrafish Tumor Model for Anticancer Evaluation of Redox-Responsive Cross-Linked Pluronic Micelles. ACS APPLIED MATERIALS & INTERFACES 2022; 14:39775-39786. [PMID: 36006680 DOI: 10.1021/acsami.2c09005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A suitable animal model for preclinical screening and evaluation in vivo could vastly increase the efficiency and success rate of nanomedicine development. Compared with rodents, the transparency of the zebrafish model offers unique advantages of real-time and high-resolution imaging of the whole body and cellular levels in vivo. In this research, we established an apoptosis-sensing xenograft zebrafish tumor model to evaluate the anti-cancer effects of redox-responsive cross-linked Pluronic polymeric micelles (CPPMs) visually and accurately. First, doxorubicin (Dox)-loaded CPPMs were fabricated and characterized with glutathione (GSH)-responsive drug release. Then, the B16F10 xenograft zebrafish tumor model was established to mimic the tumor microenvironment with angiogenesis and high GSH generation for redox-responsive tumor-targeting evaluation in vivo. The high GSH generation was first verified in the xenograft zebrafish tumor model. Compared with ordinary Pluronic polymeric micelles, Dox CPPMs had a much higher accumulation in zebrafish tumor sites. Finally, the apoptosis-sensing B16F10-C3 xenograft zebrafish tumor model was established for visual, rapid, effective, and noninvasive assessment of anti-cancer effects at the cellular level in vivo. The Dox CPPMs significantly inhibited the proliferation of cancer cells and induced apoptosis in the B16F10-C3 xenograft zebrafish tumor model. Therefore, the redox-responsive cross-linked Pluronic micelles showed effective anti-cancer therapy in the xenograft zebrafish tumor model. This xenograft zebrafish tumor model is available for rapid screening and assessment of anti-cancer effects in preclinical studies.
Collapse
Affiliation(s)
- Jinsong Tao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- Institute of Applied Physics and Materials Engineering, University of Macau, Macau 999078, China
| | - Zhengjie Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Yaxin Cheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Meng Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Qiuxia Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Wei Ge
- Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Macau 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau 999078, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- Faculty of Health Sciences, University of Macau, Macau 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau 999078, China
| |
Collapse
|
4
|
Cascallar M, Alijas S, Pensado-López A, Vázquez-Ríos AJ, Sánchez L, Piñeiro R, de la Fuente M. What Zebrafish and Nanotechnology Can Offer for Cancer Treatments in the Age of Personalized Medicine. Cancers (Basel) 2022; 14:cancers14092238. [PMID: 35565373 PMCID: PMC9099873 DOI: 10.3390/cancers14092238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer causes millions of deaths each year and thus urgently requires the development of new therapeutic strategies. Nanotechnology-based anticancer therapies are a promising approach, with several formulations already approved and in clinical use. The evaluation of these therapies requires efficient in vivo models to study their behavior and interaction with cancer cells, and to optimize their properties to ensure maximum efficacy and safety. In this way, zebrafish is an important candidate due to its high homology with the human genoma, its large offspring, and the ease in developing specific cancer models. The role of zebrafish as a model for anticancer therapy studies has been highly evidenced, allowing researchers not only to perform drug screenings but also to evaluate novel therapies such as immunotherapies and nanotherapies. Beyond that, zebrafish can be used as an “avatar” model for performing patient-derived xenografts for personalized medicine. These characteristics place zebrafish in an attractive position as a role model for evaluating novel therapies for cancer treatment, such as nanomedicine.
Collapse
Affiliation(s)
- María Cascallar
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
| | - Sandra Alijas
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Abi Judit Vázquez-Ríos
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- DIVERSA Technologies S.L., 15782 Santiago de Compostela, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
- Preclinical Animal Models Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Roberto Piñeiro
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- DIVERSA Technologies S.L., 15782 Santiago de Compostela, Spain
- Correspondence: ; Tel.: +34-981-955-704
| |
Collapse
|
5
|
Bhattacharjee S, Brayden DJ. Addressing the challenges to increase the efficiency of translating nanomedicine formulations to patients. Expert Opin Drug Discov 2020; 16:235-254. [PMID: 33108229 DOI: 10.1080/17460441.2021.1826434] [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] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Nanotechnology is in a growth phase for drug delivery and medical imaging. Nanomaterials with unique properties present opportunities for encapsulation of therapeutics and imaging agents, along with conjugation to ligands for targeting. Favorable chemistry of nanomaterials can create formulations that address critical challenges for therapeutics, such as insolubility and a low capacity to cross the blood-brain-barrier (BBB) and intestinal wall. AREAS COVERED The authors investigate challenges faced during translation of nanomedicines while suggesting reasons as to why some nanoformulations have under-performed in clinical trials. They assess physiological barriers such as the BBB and gut mucus that nanomedicines must overcome to deliver cargos. They also provide an overview with examples of how nanomedicines can be designed to improve localization and site-specific delivery (e.g., encapsulation, bioconjugation, and triggered-release). EXPERT OPINION There are examples where nanomedicines have demonstrated improved efficacy of payload in humans; however, most of the advantages conferred were in improved pharmacokinetics and reduced toxicity. Problematic data show susceptibility of nanoformulations against natural protective mechanisms present in the body, including distribution impediment by physiological barriers and activation of the reticuloendothelial system. Further initiatives should address current challenges while expanding the scope of nanomedicine into advanced biomedical imaging and antibiotic delivery.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland
| | - David J Brayden
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Belfield, Dublin, Ireland
| |
Collapse
|
6
|
Jiang F, Zhu Y, Gong C, Wei X. Atherosclerosis and Nanomedicine Potential: Current Advances and Future Opportunities. Curr Med Chem 2020; 27:3534-3554. [PMID: 30827225 DOI: 10.2174/0929867326666190301143952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 12/12/2018] [Accepted: 02/13/2019] [Indexed: 02/08/2023]
Abstract
Atherosclerosis is the leading inducement of cardiovascular diseases, which ranks the first cause of global deaths. It is an arterial disease associated with dyslipidemia and changes in the composition of the vascular wall. Besides invasive surgical strategy, the current conservative clinical treatment for atherosclerosis falls into two categories, lipid regulating-based therapy and antiinflammatory therapy. However, the existing strategies based on conventional drug delivery systems have shown limited efficacy against disease development and plenty of side effects. Nanomedicine has great potential in the development of targeted therapy, controlled drug delivery and release, the design of novel specific drugs and diagnostic modalities, and biocompatible scaffolds with multifunctional characteristics, which has led to an evolution in the diagnosis and treatment of atherosclerosis. This paper will focus on the latest nanomedicine strategies for atherosclerosis diagnosis and treatment as well as discussing the potential therapeutic targets during atherosclerosis progress, which could form the basis of development of novel nanoplatform against atherosclerosis.
Collapse
Affiliation(s)
- Fan Jiang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yunqi Zhu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xin Wei
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
7
|
Ostos FJ, Lebrón JA, López-Cornejo P, López-López M, García-Calderón M, García-Calderón CB, Rosado IV, Kalchenko VI, Rodik RV, Moyá ML. Self-aggregation in aqueous solution of amphiphilic cationic calix[4]arenes. Potential use as vectors and nanocarriers. J Mol Liq 2020. [DOI: 10.1016/j.molliq.2020.112724] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
8
|
Siemer S, Wünsch D, Khamis A, Lu Q, Scherberich A, Filippi M, Krafft MP, Hagemann J, Weiss C, Ding GB, Stauber RH, Gribko A. Nano Meets Micro-Translational Nanotechnology in Medicine: Nano-Based Applications for Early Tumor Detection and Therapy. NANOMATERIALS 2020; 10:nano10020383. [PMID: 32098406 PMCID: PMC7075286 DOI: 10.3390/nano10020383] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/03/2020] [Accepted: 02/15/2020] [Indexed: 02/07/2023]
Abstract
Nanomaterials have great potential for the prevention and treatment of cancer. Circulating tumor cells (CTCs) are cancer cells of solid tumor origin entering the peripheral blood after detachment from a primary tumor. The occurrence and circulation of CTCs are accepted as a prerequisite for the formation of metastases, which is the major cause of cancer-associated deaths. Due to their clinical significance CTCs are intensively discussed to be used as liquid biopsy for early diagnosis and prognosis of cancer. However, there are substantial challenges for the clinical use of CTCs based on their extreme rarity and heterogeneous biology. Therefore, methods for effective isolation and detection of CTCs are urgently needed. With the rapid development of nanotechnology and its wide applications in the biomedical field, researchers have designed various nano-sized systems with the capability of CTCs detection, isolation, and CTCs-targeted cancer therapy. In the present review, we summarize the underlying mechanisms of CTC-associated tumor metastasis, and give detailed information about the unique properties of CTCs that can be harnessed for their effective analytical detection and enrichment. Furthermore, we want to give an overview of representative nano-systems for CTC isolation, and highlight recent achievements in microfluidics and lab-on-a-chip technologies. We also emphasize the recent advances in nano-based CTCs-targeted cancer therapy. We conclude by critically discussing recent CTC-based nano-systems with high therapeutic and diagnostic potential as well as their biocompatibility as a practical example of applied nanotechnology.
Collapse
Affiliation(s)
- Svenja Siemer
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Désirée Wünsch
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Aya Khamis
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Qiang Lu
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Arnaud Scherberich
- Laboratory of Tissue Engineering, Universitätspital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland (M.F.)
| | - Miriam Filippi
- Laboratory of Tissue Engineering, Universitätspital Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland (M.F.)
| | - Marie Pierre Krafft
- Institut Charles Sadron (CNRS), University of Strasbourg, 23 rue du Loess, 67034 Strasbourg Cedex, France
| | - Jan Hagemann
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Carsten Weiss
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Postfach 3640, 76021 Karlsruhe, Germany
| | - Guo-Bin Ding
- Institute for Biotechnology, Shanxi University, No. 92 Wucheng Road, 030006 Taiyuan, China
| | - Roland H. Stauber
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
- Institute for Biotechnology, Shanxi University, No. 92 Wucheng Road, 030006 Taiyuan, China
- Correspondence: (R.H.S.); (A.G.); Tel.: +49-6131-176030 (A.G.)
| | - Alena Gribko
- Nanobiomedicine Department, University Medical Center Mainz/ENT, Langenbeckstrasse 1, 55131 Mainz, Germany
- Correspondence: (R.H.S.); (A.G.); Tel.: +49-6131-176030 (A.G.)
| |
Collapse
|
9
|
Lectin from Laetiporus sulphureus effectively inhibits angiogenesis and tumor development in the zebrafish xenograft models of colorectal carcinoma and melanoma. Int J Biol Macromol 2020; 148:129-139. [PMID: 31935408 DOI: 10.1016/j.ijbiomac.2020.01.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/03/2020] [Accepted: 01/04/2020] [Indexed: 12/31/2022]
Abstract
In spite of extensive usage of Laetiporus sulphureus (sulphur polypore) in traditional European and Asian ethno-medicine for centuries, its anticancer therapeutic potential and toxicity profile remained explored in animal models. Herein, using zebrafish (Danio rerio), as a preclinical animal model, we demonstrated that L. sulphureus lectin (LSL) and ethanol extract (LSE) are non-toxic at high doses up to 400-500 μg/mL, while they effectively inhibited angiogenesis and cancer development at much lower doses. Lectin showed 74-fold higher anti-angiogenic potency than the extract, and even 378-fold higher therapeutic potential than sunitinib-malate, cardiotoxic and myelosupressive anti-angiogenic drug of clinical relevance. Using wound healing and MTT assays, we proved LSL's strong antimigratory effect and selective endothelial cytotoxicity in relation to lung fibroblasts. In addition, employing the zebrafish xenograft models, we demonstrated that LSL almost completely reduced growth, neovascularization and metastasis of human colorectal carcinoma and mouse melanoma. Even more, LSL exerted 8-fold higher potency towards colorectal carcinoma than melanoma, showing markedly higher activity than cisplatin, while LSE failed to express any anticancer activity. Accompanied with non-toxic response, including neutropenia and inflammation, the results of this study strongly imply that LSL could be used as safe adjuvant in chemotherapy against colorectal carcinoma and melanoma.
Collapse
|
10
|
Tu WM, Huang XC, Chen YL, Luo YL, Liau I, Hsu HY. Longitudinal and quantitative assessment platform for concurrent analysis of anti-tumor efficacy and cardiotoxicity of nano-formulated medication in vivo. Anal Chim Acta 2019; 1095:129-137. [PMID: 31864613 DOI: 10.1016/j.aca.2019.10.019] [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] [Received: 07/11/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 01/28/2023]
Abstract
Increasing nanomedicinal approaches have been developed to effectively inhibit tumor growth; however, critical questions such as whether a nanomedicinal approach can mitigate latent side effects are barely addressed. To this end, we established a zebrafish xenograft tumor model, combining pseudodynamic three-dimensional cardiac imaging and image analysis to enable simultaneous and quantitative determination of the change of tumor volume and cardiac function of zebrafish upon specific nanoformulation treatment. Doxorubicin (DOX), a well-known chemotherapeutic agent with cardiotoxicity, and a recently developed DOX-loaded nanocomposite were employed as two model drugs to demonstrate the effectiveness to utilize the proposed evaluation platform for rapid validation. The nanoformulation significantly mitigated DOX-associated cardiotoxicity, while retaining the efficacy of DOX in inhibiting tumor growth compared to administration of carrier-free DOX at the same dose. We anticipate that this platform possesses the potential as an efficient assessment system for nanoformulated cancer therapeutics with suspected toxicity and side effects to vital organs such as the heart.
Collapse
Affiliation(s)
- Wei-Ming Tu
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao-Tung University, No.1001 Ta-Hsueh Road, Hsinchu, 30010, Taiwan, ROC
| | - Xin-Chun Huang
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao-Tung University, No.1001 Ta-Hsueh Road, Hsinchu, 30010, Taiwan, ROC
| | - Yen-Ling Chen
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao-Tung University, No.1001 Ta-Hsueh Road, Hsinchu, 30010, Taiwan, ROC
| | - Yun-Ling Luo
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao-Tung University, No.1001 Ta-Hsueh Road, Hsinchu, 30010, Taiwan, ROC
| | - Ian Liau
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao-Tung University, No.1001 Ta-Hsueh Road, Hsinchu, 30010, Taiwan, ROC; Center for Emergent Functional Matter Science, National Chiao-Tung University, Hsinchu, Taiwan.
| | - Hsin-Yun Hsu
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao-Tung University, No.1001 Ta-Hsueh Road, Hsinchu, 30010, Taiwan, ROC; Center for Emergent Functional Matter Science, National Chiao-Tung University, Hsinchu, Taiwan.
| |
Collapse
|
11
|
Xu XL, Zhu ML, Liu D, Shu GF, Qi J, Lu Y, Wang F, Ying XY, Chen J, Du YZ. Highly Integrated Nanoplatform Based on an E-Selectin-Targeting Strategy for Metastatic Breast Cancer Treatment. Mol Pharm 2019; 16:3694-3702. [PMID: 31268329 DOI: 10.1021/acs.molpharmaceut.9b00616] [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: 11/30/2022]
Abstract
Therapeutic goals for metastatic breast cancer, including shrinkage of established metastasis and suppression of movement of tumor cells, are often hard to achieve and remain the main obstacles restricting the antimetastatic efficacy of targeted drug delivery systems (TDDSs). Herein, we proposed an E-selectin-targeting nanoplatform for the systemic treatment of metastatic breast cancer. Versatile functions, including killing the circulating tumor cells, shrinking the established lesions, as well as inhibiting the movement of tumor cells, were integrated into doxorubicin-loaded sialic acid-dextran-octadecanoic acid (SDO) micelles (SDD). The prepared SDD micelles could not only inhibit lung and liver metastasis in the orthotopic 4T1 tumors model, but also decrease the metastatic lesions in the metastatic 4T1 cell model, resulting in 27.33% reduced number of metastatic nodules when compared to those without sialic acid modification. It was found that the good antimetastatic effect of SDD was only partially attributed to its ability on removing metastatic cells and metastases. Most importantly, the blank SDO micelles left in the lesion could further inhibit the cell migration and cell-cell binding. These results suggest that SA-driven TDDS has the potential for specific targeting and effective treatment of cancer metastasis.
Collapse
|
12
|
Gribko A, Künzel J, Wünsch D, Lu Q, Nagel SM, Knauer SK, Stauber RH, Ding GB. Is small smarter? Nanomaterial-based detection and elimination of circulating tumor cells: current knowledge and perspectives. Int J Nanomedicine 2019; 14:4187-4209. [PMID: 31289440 PMCID: PMC6560927 DOI: 10.2147/ijn.s198319] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cells (CTCs) are disseminated cancer cells. The occurrence and circulation of CTCs seem key for metastasis, still the major cause of cancer-associated deaths. As such, CTCs are investigated as predictive biomarkers. However, due to their rarity and heterogeneous biology, CTCs’ practical use has not made it into the clinical routine. Clearly, methods for the effective isolation and reliable detection of CTCs are urgently needed. With the development of nanotechnology, various nanosystems for CTC isolation and enrichment and CTC-targeted cancer therapy have been designed. Here, we summarize the relationship between CTCs and tumor metastasis, and describe CTCs’ unique properties hampering their effective enrichment. We comment on nanotechnology-based systems for CTC isolation and recent achievements in microfluidics and lab-on-a-chip technologies. We discuss recent advances in CTC-targeted cancer therapy exploiting the unique properties of nanomaterials. We conclude by introducing developments in CTC-directed nanosystems and other advanced technologies currently in (pre)clinical research.
Collapse
Affiliation(s)
- Alena Gribko
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Julian Künzel
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Désirée Wünsch
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Qiang Lu
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Sophie Madeleine Nagel
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Shirley K Knauer
- Department of Molecular Biology II, Center for Medical Biotechnology (ZMB)/Center for Nanointegration (CENIDE), University Duisburg-Essen, Essen 45117, Germany
| | - Roland H Stauber
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ;
| | - Guo-Bin Ding
- Nanobiomedicine Department/ENT, University Medical Center Mainz, Mainz 55131, Germany, ; .,Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, People's Republic of China,
| |
Collapse
|
13
|
Triggered doxorubicin release using redox-sensitive hyaluronic acid-g-stearic acid micelles for targeted cancer therapy. Carbohydr Polym 2019; 209:161-171. [DOI: 10.1016/j.carbpol.2019.01.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/28/2018] [Accepted: 01/07/2019] [Indexed: 12/31/2022]
|
14
|
Pan Y, Wang X, Yin Z. Synthesis and evaluation of cationic polymeric micelles as carriers of lumbrokinase for targeted thrombolysis. Asian J Pharm Sci 2019; 14:144-153. [PMID: 32104446 PMCID: PMC7032199 DOI: 10.1016/j.ajps.2018.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 12/20/2017] [Accepted: 03/26/2018] [Indexed: 02/04/2023] Open
Abstract
To achieve targeted thrombolysis, a targeted delivery system of lumbrokinase (LK) was constructed using RGDfk-conjugated hybrid micelles. Based on the specific affinity of RGDfk to glycoprotein complex of GPⅡb/Ⅲa expressed on the surface of membrane of activated platelet, LK loaded targeted micelles (LKTM) can be delivered to thrombus. The hybrid micelles were composed of polycaprolactone-block-poly (2-(dimethylamino) ethyl methacrylate) (PCL-PDMAEMA), methoxy polyethylene glycol-block- polycaprolactone (mPEG-PCL) and RGDfk conjugated polycaprolactone-block- polyethylene glycol (PCL-PEG-RGDfk). PCL-PDMAEMA was synthesized via ring open polymerization (ROP) and atom transfer radical polymerization (ATRP). PCL-PEG-RGDfk was synthesized via ROP and carbodiimide chemistry. The prepared LKTM was characterized by dynamic light scattering (DLS) and transmission electron microscope (TEM). Colloidal stability assay showed the prepared LKTM was stable. Biocompatibility assay was performed to determine the safe concentration range of polymer. The assay of fluorescent distribution in vivo demonstrated that LKTM can be efficiently delivered to thrombi in vivo. Thrombolysis in vivo indicated the thrombolytic potency of LKTM was optimal in all groups. Notably, the laboratory mice treated with LKTM exhibited a significantly shorter tail bleeding time compared to those treated with LK or LK-loaded micelles without RGDfk, which suggested that the targeted delivery of LK using RGDfk-conjugated hybrid micelles effectively reduced the bleeding risk.
Collapse
Affiliation(s)
| | | | - Zongning Yin
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
Abstract
Circulating tumor cells are a hallmark of cancer metastasis which accounts for approximately 90% of all cancer-related deaths. Their detection and characterization have significant implications in cancer biology and clinical practice. However, CTCs are rare cells and consist of heterogeneous subpopulations, requiring highly sensitive and specific techniques to identify and isolate them with high efficiency. Nanomaterials, with unique structural and functional properties, have shown strong promise to meet the challenging demands. In this review, we discuss CTC capture and therapeutic targeting, emphasizing the significance of the nanomaterials being used for this purpose. The next generation of therapy for metastatic cancer may well involve capturing and even directly neutralizing CTCs using nanomaterials.
Collapse
Affiliation(s)
- Zhenjiang Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235 USA
| | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235 USA
| |
Collapse
|
16
|
Wang X, Zhang M, Zhang L, Li L, Li S, Wang C, Su Z, Yuan Y, Pan W. Designed Synthesis of Lipid-Coated Polyacrylic Acid/Calcium Phosphate Nanoparticles as Dual pH-Responsive Drug-Delivery Vehicles for Cancer Chemotherapy. Chemistry 2017; 23:6586-6595. [DOI: 10.1002/chem.201700060] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Xin Wang
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang 110016 P. R. China
| | - Manjie Zhang
- Faculty of Chemistry; Northeast Normal University; Changchun 130024 P. R. China
| | - Lingyu Zhang
- Faculty of Chemistry; Northeast Normal University; Changchun 130024 P. R. China
| | - Lu Li
- Faculty of Chemistry; Northeast Normal University; Changchun 130024 P. R. China
| | - Shengnan Li
- Faculty of Chemistry; Northeast Normal University; Changchun 130024 P. R. China
| | - Chungang Wang
- Faculty of Chemistry; Northeast Normal University; Changchun 130024 P. R. China
| | - Zhongmin Su
- Faculty of Chemistry; Northeast Normal University; Changchun 130024 P. R. China
| | - Yue Yuan
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang 110016 P. R. China
| | - Weisan Pan
- School of Pharmacy; Shenyang Pharmaceutical University; Shenyang 110016 P. R. China
| |
Collapse
|
17
|
Gao X, Yu T, Xu G, Guo G, Liu X, Hu X, Wang X, Liu Y, Mao Q, You C, Zhou L. Enhancing the anti-glioma therapy of doxorubicin by honokiol with biodegradable self-assembling micelles through multiple evaluations. Sci Rep 2017; 7:43501. [PMID: 28240249 PMCID: PMC5327495 DOI: 10.1038/srep43501] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 01/27/2017] [Indexed: 02/05/2023] Open
Abstract
Combination chemotherapy is an important protocol in glioma therapy and honokiol shows synergistic anticancer effects with doxorubicin. In this paper, honokiol (HK) and doxorubicin (Dox) co-loaded Methoxy poly(ethylene glycol)-poly(ε-caprolactone) (MPEG-PCL) nanoparticles were prepared with a assembly method. The particle size (about 34 nm), morphology, X-ray Powder Diffraction (XRD), in vitro release profile, cytotoxicity and cell proliferation effects were studied in detail. The results indicated that honokiol and doxorubicin could be efficiently loaded into MPEG-PCL nanoparticles simultaneously, and could be released from the micelles in an extended period in vitro. In addition, honokiol and doxorubicin loaded in MPEG-PCL nanoparticles could efficiently suppress glioma cell proliferation and induce cell apoptosis in vitro. Furthermore, Dox-HK-MPEG-PCL micelles inhibited glioma growth more significantly than Dox-MPEG-PCL and HK-MPEG-PCL in both nude mice and zebrafish tumor models. Immunohistochemical analysis indicated that DOX-HK-MPEG-PCL micelles improved Dox's anti-tumor effect by enhancing tumor cell apoptosis, suppressing tumor cell proliferation, and inhibiting angiogenesis. Our data suggest that Dox-HK-MPEG-PCL micelles have the potential to be applied clinically in glioma therapy.
Collapse
Affiliation(s)
- Xiang Gao
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
- Department of Pharmacology, Yale School of Medicine, Yale University, New Haven, Connecticut, 06520, USA
- Institute of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Ting Yu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
| | - Guangya Xu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
| | - Gang Guo
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
| | - Xiaoxiao Liu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
| | - Xin Hu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
| | - Xiang Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
- Institute of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Yanhui Liu
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
| | - Qing Mao
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
| | - Chao You
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
- Institute of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Liangxue Zhou
- Department of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation center, Chengdu, 610041, PR China
| |
Collapse
|
18
|
Chen N, Qin S, Yang X, Wang Q, Huang J, Wang K. "Sense-and-Treat" DNA Nanodevice for Synergetic Destruction of Circulating Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2016; 8:26552-26558. [PMID: 27653943 DOI: 10.1021/acsami.6b08695] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
DNA nanostrucures are promising materials for biomedical applications. Herein, we established a "sense-and-treat" localized drug delivery system based on a DNA nanodevice to specifically destroy circulating tumor cells (CTCs) by synergetic chemotherapy and photodynamic therapy. The DNA nanodevices could sense the existence of CTCs and treat CTCs with anticancer agents. Typically, the presence of target cell promoted the formation of hairpin structure of aptamer, and then the aptamer-accompanied DNA tetrahedron would release from the supporter. The chemotherapy drugs (doxorubicin, Dox) loaded in DNA tetrahedron would destroy the CTCs specifically. Moreover, the photosensitizer labeled on DNA tetrahedron would be activated by lights and generated toxic 1O2, once DNA nanodevices bound CTCs flow through the superficial capillary. Unlike the aptamer only labeled with photosensitizer, the DNA nanodevice showed the capability to promote cellular internalization of anticancer agents, increase drug loading capacity, and realize synergetic therapy, which enhanced the destructive ability of anticancer agents. As proof of concept, this DNA nanodevice has the potential to inhibit metastasis by synergetic destruction of CTCs.
Collapse
Affiliation(s)
- Nandi Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University , Changsha 410082, China
| | - Shiya Qin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University , Changsha 410082, China
| | - Xiaohai Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University , Changsha 410082, China
| | - Qing Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University , Changsha 410082, China
| | - Jin Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University , Changsha 410082, China
| | - Kemin Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University , Changsha 410082, China
| |
Collapse
|
19
|
Chen N, Yang X, Wang Q, Jian L, Shi H, Qin S, Wang K, Huang J, Liu W. Proof of concept for inhibiting metastasis: circulating tumor cell-triggered localized release of anticancer agent via a structure-switching aptamer. Chem Commun (Camb) 2016; 52:6789-92. [PMID: 27121864 DOI: 10.1039/c6cc02374f] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Existing drug delivery systems were not suitable for killing cells in the circulatory system specifically. Herein, we developed a novel localized drug delivery strategy, in which the release of anticancer agents was specifically triggered by circulating tumor cells. Meanwhile, damage to non-target cells was avoided.
Collapse
Affiliation(s)
- Nandi Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Huang XC, Wu LB, Hsu JF, Shigeto S, Hsu HY. Biothiol-triggered, self-disassembled silica nanobeads for intracellular drug delivery. Acta Biomater 2015; 23:263-270. [PMID: 25983312 DOI: 10.1016/j.actbio.2015.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 05/04/2015] [Accepted: 05/10/2015] [Indexed: 10/23/2022]
Abstract
Silica-based nanomaterials have demonstrated great potential in biomedical applications due to their chemical inertness. However, the degradability and endosomal trapping issues remain as rate-limiting barriers during their innovation. In this study, we provide a simple yet novel sol-gel approach to construct the redox-responsive silica nanobeads (ReSiNs), which could be rapidly disassembled upon redox gradient for intracellular drug delivery. The disulfide-linked scaffold of the nanobead was synthesized by employing the dithiobis-(succinimidyl propionate) to bridge (3-aminopropyl)-trimethoxysilane. Such silica matrix could be efficiently disrupted in response to intracellular glutathione, resulting in drug release and collapse of entire nanocarrier. Moreover, the ReSiNs exhibited insignificant cytotoxicity before and after the degradation. These results indicated the potential of using ReSiNs as a novel silica-based, biothiol-degradable nanoplatform for future drug delivery.
Collapse
|