101
|
Zhao P, Li M, Wang Y, Chen Y, He C, Zhang X, Yang T, Lu Y, You J, Lee RJ, Xiang G. Enhancing anti-tumor efficiency in hepatocellular carcinoma through the autophagy inhibition by miR-375/sorafenib in lipid-coated calcium carbonate nanoparticles. Acta Biomater 2018; 72:248-255. [PMID: 29555460 DOI: 10.1016/j.actbio.2018.03.022] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/23/2018] [Accepted: 03/07/2018] [Indexed: 12/19/2022]
Abstract
Sorafenib is a first-line drug for hepatocellular carcinoma (HCC). Autophagy has been shown to facilitate sorafenib resistance. miR-375 has been shown to be an inhibitor of autophagy. In this study, miR-375 and sorafenib were co-loaded into calcium carbonate nanoparticles with lipid coating (miR-375/Sf-LCC NPs). The nanoparticles had high loading efficiency and were ∼50 nm in diameter. Besides, the NPs could increase the stability and residence time of both drugs. Moreover, we demonstrated that autophagy was activated in HCC cells by sorafenib but not by miR-375/Sf-LCC NPs. In vitro, miR-375/Sf-LCC NPs exhibited pH-dependent drug release and potent cytotoxicity. In vivo, miR-375/Sf-LCC NPs increased miR-375 and sorafenib uptake in tumor (2 folds compared with Lipofectamine 2000-miR-375 and 2-5 folds compared with free sorafenib). Furthermore, miR-375/Sf-LCC NPs showed greatly enhanced therapeutic efficacy in an HCC xenograft model. These findings suggest that miR-375/Sf-LCC NPs may be a promising agent for the HCC therapy. STATEMENT OF SIGNIFICANCE Hepatocellular carcinoma (HCC) is the most common primary liver tumor and the third leading cause of cancer mortality globally. In this manuscript, miR-375 and sorafenib were co-loaded into calcium carbonate nanoparticles with lipid coating (miR-375/Sf-LCC NPs) to treat HCC. We demonstrated that miR-375/Sf-LCC NPs can deliver sorafenib and miR-375 into HCC cells and tumor tissues, increase drug retention time in tumor, significantly inhibit autophagy and produce enhanced anti-tumor effect.
Collapse
|
102
|
Jin M, Jin G, Kang L, Chen L, Gao Z, Huang W. Smart polymeric nanoparticles with pH-responsive and PEG-detachable properties for co-delivering paclitaxel and survivin siRNA to enhance antitumor outcomes. Int J Nanomedicine 2018; 13:2405-2426. [PMID: 29719390 PMCID: PMC5916383 DOI: 10.2147/ijn.s161426] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The co-delivery of chemotherapeutic agents and small interfering RNA (siRNA) within one cargo can enhance the anticancer outcomes through its synergistic therapeutic effects. Materials and methods We prepared smart polymeric nanoparticles (NPs) with pH-responsive and poly(ethylene glycol) (PEG)-detachable properties to systemically co-deliver paclitaxel (PTX) and siRNA against survivin gene for lung cancer therapy. The cationic polyethyleneimine-block-polylactic acid (PEI-PLA) was first synthesized and characterized, with good biocompatibility. PTX was encapsulated into the hydrophobic core of the PEI-PLA polymers by dialysis, and then the survivin siRNA was loaded onto the PTX-loaded NPs (PEI-PLA/PTX) through electrostatic interaction between siRNA and PEI block. Finally, the negatively charged poly(ethylene glycol)-block-poly(L-aspartic acid sodium salt) (PEG-PAsp) was coated onto the surface of NPs by electrostatic interaction to form final smart polymeric NPs with mean particle size of 82.4 nm and zeta potential of 4.1 mV. After uptake of NPs by tumor cells, the PEG-PAsp segments became electrically neutral owing to the lower endosome pH and consequently detached from the smart NPs. This process allowed endosomal escape of the NPs through the proton-sponge effect of the exposed PEI moiety. Results The resulting NPs achieved drug loading of 6.04 wt% and exhibited good dispersibility within 24 h in 10% fetal bovine serum (FBS). At pH 5.5, the NPs presented better drug release and cellular uptake than at pH 7.4. The NPs with survivin siRNA effectively knocked down the expression of survivin mRNA and protein owing to enhanced cell uptake of NPs. Cell counting kit-8 (CCK-8) assay showed that the NPs presented low systemic toxicity and improved antiproliferation effect of PTX on A549 cells. Moreover, in vivo studies demonstrated that accumulated NPs in the tumor site were capable of inhibiting the tumor growth and extending the survival rate of the mice by silencing the survivin gene and delivering PTX into tumor cells simultaneously. Conclusion These results indicate that the prepared nano-vectors could be a promising co-delivery system for novel chemo/gene combination therapy.
Collapse
Affiliation(s)
- Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangming Jin
- Department of Diagnostic Radiology 2, Yanbian University Hospital, Yanji, Jilin, China
| | - Lin Kang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
103
|
Pugliese E, Coentro JQ, Zeugolis DI. Advancements and Challenges in Multidomain Multicargo Delivery Vehicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1704324. [PMID: 29446161 DOI: 10.1002/adma.201704324] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/05/2017] [Indexed: 06/08/2023]
Abstract
Reparative and regenerative processes are well-orchestrated temporal and spatial events that are governed by multiple cells, molecules, signaling pathways, and interactions thereof. Yet again, currently available implantable devices fail largely to recapitulate nature's complexity and sophistication in this regard. Herein, success stories and challenges in the field of layer-by-layer, composite, self-assembly, and core-shell technologies are discussed for the development of multidomain/multicargo delivery vehicles.
Collapse
Affiliation(s)
- Eugenia Pugliese
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| | - João Q Coentro
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| |
Collapse
|
104
|
Li T, Shen X, Xie X, Chen Z, Li S, Qin X, Yang H, Wu C, Liu Y. Irinotecan/IR-820 coloaded nanocomposite as a cooperative nanoplatform for combinational therapy of tumor. Nanomedicine (Lond) 2018; 13:595-603. [DOI: 10.2217/nnm-2017-0315] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Aim: To enhance synergistic therapeutic effects in breast cancer therapy. Here, we used hollow mesoporous silica nanoparticles as a biocompatible carrier to coload chemotherapy drugs Irinotecan and near-infrared IR-820 dye, which enhanced antitumor efficacy by combining chemotherapy and phototherapy. Methods: The successful synthesis of hollow mesoporous silica nanoparticles/Irinotecan/IR820 (HMII) nanocomplex was confirmed by Fourier transform infrared spectroscopy and Fluorescence spectra. The photothermal conversion efficiency and antitumor efficiency in murine breast cancer cells (EMT-6) bearing mice were further evaluated. Results: The results demonstrated that HMII enhanced the delivery of Irinotecan and IR-820 into EMT-6 cells. HMII generated a high temperature upon a near-infrared laser irradiation (808 nm), and showed higher therapeutic efficacy in EMT-6-bearing mice compared with either HMII without laser or free drug with a laser. Conclusion: HMII is a desired drug codelivery system to efficiently inhibit the growth of breast cancer.
Collapse
Affiliation(s)
- Tingting Li
- Department of Biophysics, School of Life Science & Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xue Shen
- Department of Biophysics, School of Life Science & Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiaoxue Xie
- Department of Biophysics, School of Life Science & Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Zhongyuan Chen
- Department of Biophysics, School of Life Science & Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Shun Li
- Department of Biophysics, School of Life Science & Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
- Center for Information in Biology, University of Electronic Science & Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiang Qin
- Department of Biophysics, School of Life Science & Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
- Center for Information in Biology, University of Electronic Science & Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hong Yang
- Department of Biophysics, School of Life Science & Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
- Center for Information in Biology, University of Electronic Science & Technology of China, Chengdu 610054, Sichuan, PR China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science & Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
- Center for Information in Biology, University of Electronic Science & Technology of China, Chengdu 610054, Sichuan, PR China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science & Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
- Center for Information in Biology, University of Electronic Science & Technology of China, Chengdu 610054, Sichuan, PR China
| |
Collapse
|
105
|
Sequential delivery of therapeutic agents using a rationally designed disulfide-linked glycolipid-like nanocarrier. Oncotarget 2018; 7:83258-83269. [PMID: 27825127 PMCID: PMC5347767 DOI: 10.18632/oncotarget.13083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 10/13/2016] [Indexed: 01/08/2023] Open
Abstract
Usage of combination therapies to deliver multiple therapeutics to increase treatment efficacy has shown promising results in the clinic. In an effort to maximize the synergistic effect of co-delivery of a drug and siRNA, we have developed a time-dependent sequential drug delivery system (DDS) based on a disulfide-linked chitosan-based nanocarrier (CS-ss-SA) for the co-delivery of paclitaxel (PTX) and Bcl-2 specific siRNA (siBcl-2). This CS-ss-SA nanocarrier is able to transport both drug and siRNA by entrapment of PTX and adsorption of siRNA on the shell by electrostatic attraction. We show that this nanocarrier transports siRNA into tumor cells via its glycolipid-like spatial structure and releases a hydrophobic model drug, Nile Red 8-11 h later. Next, when siRNA and the hydrophobic drug PTX were co-delivered to tumor cells, a synergistic effect was observed in both cell cycle arrest and cell viability. Ultimately, the co-delivery of PTX and siBcl-2 by CS-ss-SA may prove to be more efficacious and may even help overcome drug resistance.
Collapse
|
106
|
Kong N, Deng M, Sun XN, Chen YD, Sui XB. Polydopamine-Functionalized CA-(PCL- ran-PLA) Nanoparticles for Target Delivery of Docetaxel and Chemo-photothermal Therapy of Breast Cancer. Front Pharmacol 2018; 9:125. [PMID: 29527167 PMCID: PMC5829531 DOI: 10.3389/fphar.2018.00125] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 02/05/2018] [Indexed: 01/04/2023] Open
Abstract
Current limitations of cancer therapy include the lack of effective strategy for target delivery of chemotherapeutic drugs, and the difficulty of achieving significant efficacy by single treatment. Herein, we reported a synergistic chemo-photothermal strategy based on aptamer (Apt)-polydopamine (pD) functionalized CA-(PCL-ran-PLA) nanoparticles (NPs) for effective delivery of docetaxel (DTX) and enhanced therapeutic effect. The developed DTX-loaded Apt-pD-CA-(PCL-ran-PLA) NPs achieved promising advantages, such as (i) improved drug loading content (LC) and encapsulation efficiency (EE) initiated by star-shaped copolymer CA-(PCL-ran-PLA); (ii) effective target delivery of drugs to tumor sites by incorporating AS1411 aptamers; (iii) significant therapeutic efficacy caused by synergistic chemo-photothermal treatment. In addition, the pD coating strategy with simple procedures could address the contradiction between targeting modification and maintaining formerly excellent bio-properties. Therefore, with excellent bio-properties and simple preparation procedures, the DTX-loaded Apt-pD-CA-(PCL-ran-PLA) NPs effectively increased the local drug concentration in tumor sites, minimized side effects, and significantly eliminated tumors, indicating the promising application of these NPs for cancer therapy.
Collapse
Affiliation(s)
- Na Kong
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Mei Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiu-Na Sun
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Yi-Ding Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin-Bing Sui
- Department of Medical Oncology, Holistic Integrative Oncology Institutes and Holistic Integrative Cancer Center of Traditional Chinese and Western Medicine, The Affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, China
- Department of Cancer Pharmacology, Holistic Integrative Pharmacy Institutes, College of Medicine, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
107
|
Wu T, Zhang D, Qiao Q, Qin X, Yang C, Kong M, Deng H, Zhang Z. Biomimetic Nanovesicles for Enhanced Antitumor Activity of Combinational Photothermal and Chemotherapy. Mol Pharm 2018; 15:1341-1352. [PMID: 29397741 DOI: 10.1021/acs.molpharmaceut.7b01142] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The combination of multiple modalities has shown great potential in cancer treatment with improved therapeutic effects and minimized side effects. Here, we fabricated a type of doxorubicin-encapsulated biomimetic nanovesicle (NV) by a facile method with near-infrared dye insertion in the membrane for combinatorial photothermal and chemotherapy. With innate biomimetic properties, NVs enhanced the uptake by tumor cells while reducing the phagocytosis of macrophages. Upon laser irradiation, NVs can convert the absorbed fluorescent energy into heat for effective tumor killing. Hyperthermia can further induce membrane ablation of NVs to accelerate the release of chemotherapeutic drug for potent cytotoxicity to tumor cells. The NVs improved drug accumulation and showed a more efficient in vivo photothermal effect with a rapid temperature increase in tumors. Moreover, the NV-based combinational photothermal and chemotherapy exhibited significant tumor growth suppression with a high inhibitory rate of 91.6% and negligible systemic toxicity. The results indicate that NVs could be an appealing vehicle for combinational cancer treatment.
Collapse
|
108
|
Bauer KN, Liu L, Andrienko D, Wagner M, Macdonald EK, Shaver MP, Wurm FR. Polymerizing Phostones: A Fast Way to In-Chain Poly(phosphonate)s with Adjustable Hydrophilicity. Macromolecules 2018. [DOI: 10.1021/acs.macromol.7b02473] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Kristin N. Bauer
- Max-Planck-Institut
für Polymerforschunng, Ackermannweg
10, 55128 Mainz, Germany
| | - Lei Liu
- Max-Planck-Institut
für Polymerforschunng, Ackermannweg
10, 55128 Mainz, Germany
| | - Denis Andrienko
- Max-Planck-Institut
für Polymerforschunng, Ackermannweg
10, 55128 Mainz, Germany
| | - Manfred Wagner
- Max-Planck-Institut
für Polymerforschunng, Ackermannweg
10, 55128 Mainz, Germany
| | - Emily K. Macdonald
- School
of Chemistry, University of Edinburgh, Joseph Black Building, David Brewster
Road, Edinburgh EH9 3FJ, United Kingdom
| | - Michael P. Shaver
- School
of Chemistry, University of Edinburgh, Joseph Black Building, David Brewster
Road, Edinburgh EH9 3FJ, United Kingdom
| | - Frederik R. Wurm
- Max-Planck-Institut
für Polymerforschunng, Ackermannweg
10, 55128 Mainz, Germany
| |
Collapse
|
109
|
Jiang Y, Reineke TM, Lodge TP. Complexation of DNA with Cationic Copolymer Micelles: Effects of DNA Length and Topology. Macromolecules 2018. [DOI: 10.1021/acs.macromol.7b02201] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Yaming Jiang
- Department of Chemical Engineering & Materials Science and ‡Department of Chemistry, University of Minnesota, 421 Washington Ave. SE, Minneapolis, Minnesota 55455, United States
| | - Theresa M. Reineke
- Department of Chemical Engineering & Materials Science and ‡Department of Chemistry, University of Minnesota, 421 Washington Ave. SE, Minneapolis, Minnesota 55455, United States
| | - Timothy P. Lodge
- Department of Chemical Engineering & Materials Science and ‡Department of Chemistry, University of Minnesota, 421 Washington Ave. SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
110
|
Krivitsky A, Polyak D, Scomparin A, Eliyahu S, Ofek P, Tiram G, Kalinski H, Avkin-Nachum S, Feiner Gracia N, Albertazzi L, Satchi-Fainaro R. Amphiphilic poly(α)glutamate polymeric micelles for systemic administration of siRNA to tumors. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2018; 14:303-315. [PMID: 29127036 DOI: 10.1016/j.nano.2017.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/08/2017] [Accepted: 10/30/2017] [Indexed: 01/27/2023]
Abstract
RNAi therapeutics carried a great promise to the area of personalized medicine: the ability to target "undruggable" oncogenic pathways. Nevertheless, their efficient tumor targeting via systemic administration had not been resolved yet. Amphiphilic alkylated poly(α)glutamate amine (APA) can serve as a cationic carrier to the negatively-charged oligonucleotides. APA polymers complexed with siRNA to form round-shaped, homogenous and reproducible nano-sized polyplexes bearing ~50 nm size and slightly negative charge. In addition, APA:siRNA polyplexes were shown to be potent gene regulators in vitro. In light of these preferred physico-chemical characteristics, their performance as systemically-administered siRNA nanocarriers was investigated. Intravenously-injected APA:siRNA polyplexes accumulated selectively in tumors and did not accumulate in the lungs, heart, liver or spleen. Nevertheless, the polyplexes failed to induce specific mRNA degradation, hence neither reduction in tumor volume nor prolonged mice survival was seen.
Collapse
Affiliation(s)
- Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Dina Polyak
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Anna Scomparin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Shay Eliyahu
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
111
|
Sheena TS, Balaji P, Venkatesan R, Akbarsha MA, Jeganathan K. Functional evaluation of doxorubicin decorated polymeric liposomal curcumin: a surface tailored therapeutic platform for combination chemotherapy. NEW J CHEM 2018. [DOI: 10.1039/c8nj02406e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The present study deals with the hypothesis and design of new platform for the accommodation of curcumin and doxorubicin in surface engineered liposomes for combination chemotherapy.
Collapse
Affiliation(s)
- Thankaraj Salammal Sheena
- Centre for Nanoscience and Nanotechnology
- Department of Physics
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| | - Perumalsamy Balaji
- National Centre for Alternatives to Animal Experiments (NCAAE)
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| | - Rajiu Venkatesan
- Centre for Nanoscience and Nanotechnology
- Department of Physics
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| | | | - K. Jeganathan
- Centre for Nanoscience and Nanotechnology
- Department of Physics
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| |
Collapse
|
112
|
Nehate C, Moothedathu Raynold AA, Koul V. ATRP Fabricated and Short Chain Polyethylenimine Grafted Redox Sensitive Polymeric Nanoparticles for Codelivery of Anticancer Drug and siRNA in Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:39672-39687. [PMID: 29048878 DOI: 10.1021/acsami.7b11716] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
To overcome the limitations of conventional chemotherapy, nanoparticle-mediated combinatorial delivery of siRNA and drugs represents a new approach to overcome its associated side effects. Designing safe and efficient vehicles for their codelivery has emerged as a potential challenge in the clinical translation of these formulations. Herein, we have demonstrated a novel "two-in-one" polyplex nanosystem developed from redox sensitive, short chain polyethylenimine modified poly[(poly(ethylene)glycol methacrylate]-s-s-polycaprolactone copolymer synthesized by atom-transfer free-radical polymerization (ATRP), which can deliver doxorubicin and polo-like kinase I (plk1) siRNA, simultaneously for an enhanced chemotherapeutic effect. The nanoparticles were found to be stable at physiological buffer with and without fetal bovine serum (FBS). The developed polymeric nanosystem was found to be biocompatible and hemocompatible in vitro and in vivo at repeated dose administrations. The polymer could easily self-assemble into ∼100 nm spherical nanoparticles with enhanced doxorubicin loading (∼18%) and effective siRNA complexation at a polymer to siRNA weight ratio of 15. The doxorubicin loaded nanoparticles exhibited ∼4-fold higher drug release in endosomal pH (pH 5) containing 10 mmol of GSH compared to pH 7.4, depicting their redox-sensitive behavior. The polyplexes were capable of delivering both cargos simultaneously to cancer cells in vitro as observed by their excellent colocalization in the cytoplasm of MDA-MB-231 and HeLa cells using confocal laser microscopy. Moreover, in vitro transfection of the cells with polyplexes exhibited 50-70% knockdown of plk1-mRNA expression in both cell lines. In vivo administration of the drug loaded polyplexes to EAT tumor bearing (EAT, Ehrlich ascites tumor) Swiss albino mice showed a ∼29-fold decrease in percent tumor volume in comparison to the control group. The results highlight the therapeutic potential of the polyplexes as a combined delivery of doxorubicin and plk1-siRNA in cancer therapy.
Collapse
Affiliation(s)
- Chetan Nehate
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi , New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences , New Delhi 110029, India
| | - Aji Alex Moothedathu Raynold
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi , New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences , New Delhi 110029, India
| | - Veena Koul
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi , New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences , New Delhi 110029, India
| |
Collapse
|
113
|
Yu C, Ding B, Zhang X, Deng X, Deng K, Cheng Z, Xing B, Jin D, Ma P, Lin J. Targeted iron nanoparticles with platinum-(IV) prodrugs and anti-EZH2 siRNA show great synergy in combating drug resistance in vitro and in vivo. Biomaterials 2017; 155:112-123. [PMID: 29175080 DOI: 10.1016/j.biomaterials.2017.11.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 10/21/2017] [Accepted: 11/13/2017] [Indexed: 01/07/2023]
Abstract
Resistance to platinum agents is challenging in cancer treatment with platinum drugs. Such resistant cells prevent effective platinum accumulation intracellular and alter cellular adaptations to survive from cytotoxicity by regulating corresponding proteins expression. Ideal therapeutics should combine resolution to these pump and non-pump relevant resistance of cancer cells to achieve high efficacy and low side effect. Fe3O4 nanocarrier loaded with drugs could enter cells in a more efficient endocytosis manner which circumvents pump-relevant drug resistance. EZH2 protein which was previously found to be over-expressed in drug-resistant cancer cells was reported to be involved in platinum drug resistance and play a vital role in anti-apoptosis pathways. Here, we report Fe3O4 nanoparticles loaded with siEZH2 (siRNA), a platinum prodrug in +4 oxidation state (cis, cis, trans-diamminedichlorodisuccinato-platinum-(IV), namely Pt(IV)) and luteinizing hormone-releasing hormone (LHRH) targeting polypeptides. Results show that targeted nanoparticles loading with siEZH2 synergize with Pt(IV) and result in similar cell killing performance to A2780/DDP cells (cisplatin resistant) compared with non-siEZH2 loaded nanoparticles to A2780 cells (cisplatin sensitive). Thus, this Fe3O4@PEI-Pt(IV)-PEG-LHRH@siEZH2 nanoparticles reverse the cisplatin resistance from the pump and non-pump relevant aspects, fully taking advantage of nanocarrier system.
Collapse
Affiliation(s)
- Chang Yu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China
| | - Xinyang Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Xiaoran Deng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kerong Deng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ziyong Cheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Bengang Xing
- School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore
| | - Dayong Jin
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW, 2007, Australia
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.
| |
Collapse
|
114
|
Cheng W, Liang C, Wang X, Tsai HI, Liu G, Peng Y, Nie J, Huang L, Mei L, Zeng X. A drug-self-gated and tumor microenvironment-responsive mesoporous silica vehicle: "four-in-one" versatile nanomedicine for targeted multidrug-resistant cancer therapy. NANOSCALE 2017; 9:17063-17073. [PMID: 29085938 DOI: 10.1039/c7nr05450e] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The design of multifunctional nanocarriers for the co-delivery of anticancer drugs and genetic agents offers an effective and promising strategy to combat multidrug-resistant cancer. Herein, we developed a simple and facile method to fabricate a drug-self-gated and pH-sensitive mesoporous silica vehicle as a "four-in-one" versatile co-delivery system, which possesses targeted chemo and gene therapy capability against multidrug-resistant cancer. P-gp siRNA molecules were loaded into the channels of mesoporous silica nanoparticles. A chemotherapeutic drug (DOX) was employed as a gatekeeper via a pH-sensitive benzoic-imine covalent bond. Folic acid conjugation onto the surface endowed this system with an excellent tumor-targeting effect, which was demonstrated by the cellular and tumor targeting assay. The effective downregulation of P-gp protein by the co-delivered P-gp siRNA was observed by western blotting. Both the in vitro cell viability study and in vivo tumor inhibition assay showed a synergistic effect in suppressing cancer cell proliferation. Therefore, this drug-self-gated nanosystem exhibited great potential for improved multidrug-resistant cancer treatment without any further potential risks of capping agents.
Collapse
Affiliation(s)
- Wei Cheng
- The Shenzhen Key Lab of Gene and Antibody Therapy, The Ministry-Province Jointly Constructed Base for State Key Lab-Shenzhen Key Laboratory of Chemical Biology, and Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Long J, Yang Y, Kang T, Zhao W, Cheng H, Wu Y, Du T, Liu B, Li Y, Luo F, Gou M. Ovarian Cancer Therapy by VSVMP Gene Mediated by a Paclitaxel-Enhanced Nanoparticle. ACS APPLIED MATERIALS & INTERFACES 2017; 9:39152-39164. [PMID: 28944654 DOI: 10.1021/acsami.7b10796] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Jianlin Long
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Department of Medical Oncology, Lung Cancer Center, Cancer Center,
West China Hospital, Medical School, Sichuan University, Chengdu, Sichuan 610041, P. R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
- Department of Oncology, West China Guang’an Hospital, Sichuan University, Guang’an, Sichuan 638000, P. R. China
| | - Yuping Yang
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| | - Tianyi Kang
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| | - Wei Zhao
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| | - Hao Cheng
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| | - Yujiao Wu
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| | - Ting Du
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| | - Beibei Liu
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| | - Yang Li
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| | - Feng Luo
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Department of Medical Oncology, Lung Cancer Center, Cancer Center,
West China Hospital, Medical School, Sichuan University, Chengdu, Sichuan 610041, P. R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| | - Maling Gou
- State Key Laboratory of Biotherapy
and cancer center, West China Hospital, Sichuan University, and Collaborative
Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
- Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
116
|
Ruttala HB, Chitrapriya N, Kaliraj K, Ramasamy T, Shin WH, Jeong JH, Kim JR, Ku SK, Choi HG, Yong CS, Kim JO. Facile construction of bioreducible crosslinked polypeptide micelles for enhanced cancer combination therapy. Acta Biomater 2017; 63:135-149. [PMID: 28890258 DOI: 10.1016/j.actbio.2017.09.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 08/14/2017] [Accepted: 09/02/2017] [Indexed: 12/27/2022]
Abstract
In this study, we developed pH and redox-responsive crosslinked polypeptide-based combination micelles for enhanced chemotherapeutic efficacy and minimized side effects. The stability and drug release properties of the polypeptide micelles were efficiency balanced by the corona-crosslinking of the triblock copolymer, poly(ethylene glycol)-b-poly(aspartic acid)-b-poly(tyrosine) (PEG-b-pAsp-b-pTyr) with coordinated redox and pH dual-sensitivity by introducing disulfide crosslinkages. Because of the crosslinking of the middle shell of the triblock polypeptide micelles, their robust structure was maintained in strong destabilization conditions and exhibited excellent stability. GSH concentrations were significantly higher in tumor tissue than in normal tissue, which formed the basis for our design. Drug release was elevated under redox and low acidic conditions. Furthermore, crosslinked micelles showed a superior anticancer effect compared to that of non-crosslinked micelles. Incorporation of docetaxel (DTX) and lonidamine (LND) in crosslinked polypeptide micelles increased the intracellular reactive oxygen species (ROS) level and oxidative stress and caused damage to intracellular components that resulted in greater apoptosis of cancer cells than when DTX or LND was used alone. The combination of DTX and LND in crosslinked micelles exhibited efficacious inhibition of tumor growth with an excellent safety profile compared to that reported for drug cocktail combinations and non-crosslinked micelles. Overall, redox/pH-responsive polypeptide micelles could be an interesting platform for efficient chemotherapy. STATEMENT OF SIGNIFICANCE We have synthesized a biodegradable polypeptide block copolymer to construct a facile pH and redox-responsive polymeric micelle asan advanced therapeutic system for cancer therapy. We have designed a corona-crosslinked triblock copolymer (poly (ethylene glycol)-b-poly(aspartic acid)-b-poly(tyrosine) (PEG-b-pAsp-b-pTyr)) micelles co-loaded with docetaxel and lonidamine (cl-M/DL). The corona of triblock polymer was crosslinked to maintain its structural integrity in the physiological environment. The mitochondrial targeting LND is expected to generate ROS, oxidative stress and thereby synergize the chemotherapeutic efficacy of DTX in killing cancer cells. Consistently, cl-M/DL exhibited excellent antitumor efficacy in xenograft tumor model with remarkable tumor regression. Overall, we demonstrated the construction of bioreducible nanosystem for the effective synergistic delivery of DTX/LND in tumor tissues towards cancer treatment.
Collapse
|
117
|
Zhao P, Wu S, Cheng Y, You J, Chen Y, Li M, He C, Zhang X, Yang T, Lu Y, Lee RJ, He X, Xiang G. MiR-375 delivered by lipid-coated doxorubicin-calcium carbonate nanoparticles overcomes chemoresistance in hepatocellular carcinoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2507-2516. [DOI: 10.1016/j.nano.2017.05.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/06/2017] [Accepted: 05/18/2017] [Indexed: 12/21/2022]
|
118
|
Da Silva CG, Peters GJ, Ossendorp F, Cruz LJ. The potential of multi-compound nanoparticles to bypass drug resistance in cancer. Cancer Chemother Pharmacol 2017; 80:881-894. [PMID: 28887666 PMCID: PMC5676819 DOI: 10.1007/s00280-017-3427-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/29/2017] [Indexed: 01/28/2023]
Abstract
PURPOSE The therapeutic efficacy of conventional chemotherapy against several solid tumors is generally limited and this is often due to the development of resistance or poor delivery of the drugs to the tumor. Mechanisms of resistance may vary between cancer types. However, with current development of genetic analyses, imaging, and novel delivery systems, we may be able to characterize and bypass resistance, e.g., by inhibition of the right target at the tumor site. Therefore, combined drug treatments, where one drug will revert or obstruct the development of resistance and the other will concurrently kill the cancer cell, are rational solutions. However, drug exposure of one drug will defer greatly from the other due to their physicochemical properties. In this sense, multi-compound nanoparticles are an excellent modality to equalize drug exposure, i.e., one common physicochemical profile. In this review, we will discuss novel approaches that employ nanoparticle technology that addresses specific mechanisms of resistance in cancer. METHODS The PubMed literature was consulted and reviewed. RESULTS Nanoparticle technology is emerging as a dexterous solution that may address several forms of resistance in cancer. For instance, we discuss advances that address mechanisms of resistance with multi-compound nanoparticles which co-deliver chemotherapeutics with an anti-resistance agent. Promising anti-resistance agents are (1) targeted in vivo gene silencing methods aimed to disrupt key resistance gene expression or (2) protein kinase inhibitors to disrupt key resistance pathways or (3) efflux pumps inhibitors to limit drug cellular efflux.
Collapse
Affiliation(s)
- C G Da Silva
- Translational Nanobiomaterials and Imaging, Department of Radiology, Bldg.1, C2-187h, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, Leiden, The Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Bldg.1, C2-187h, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
119
|
Multifunctional nanoparticles co-delivering EZH2 siRNA and etoposide for synergistic therapy of orthotopic non-small-cell lung tumor. J Control Release 2017; 268:198-211. [PMID: 29061511 DOI: 10.1016/j.jconrel.2017.10.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 05/17/2017] [Accepted: 10/14/2017] [Indexed: 12/29/2022]
Abstract
Malignant proliferation and metastasis in non-small cell lung carcinoma (NSCLC) are great challenges for effective clinical treatment through conventional chemotherapy. The combinational therapy strategy of RNA interfering (RNAi) technology and chemotherapeutic agents have been reported to be promising for effective cancer therapy. In this study, based on multifunctional nanoparticles (NPs), the simultaneous delivery of etoposide (ETP) and anti-Enhancer of Zeste Homologue 2 (EZH2) siRNA for the effective treatment of orthotopic lung tumor was achieved. The NPs exhibited pH/redox dual sensitivity verified by particle size changes, morphological changes, and in vitro release of drugs. Confocal microscopy analysis confirmed that the NPs exhibited endosomal escape property and on-demand intracellular drug release behavior, which can protect siRNA from degradation and facilitate the chemotherapeutic effect respectively. In vitro tumor cell motility study demonstrated that EZH2 siRNA loaded in NPs can decrease the migration and invasion capabilities of tumor cells by downregulating the expression of EZH2 mRNA and protein. In particular, an antiproliferation study revealed that the co-delivery of siRNA and ETP in the multifunctional NPs can induce a synergistic therapeutic effect on NSCLC. In vivo targeting evaluation showed that cRGDyC-PEG modification on NPs exhibited a low distribution in normal organs and an obvious accumulation in orthotopic lung tumor. Furthermore, targeted NPs co-delivering siRNA and ETP showed superior inhibition on tumor growth and metastasis and produced minimal systemic toxicity. These findings indicated that multifunctional NPs can be utilized as a co-delivery system, and that the combination of EZH2 siRNA and ETP can effectively treat NSCLC.
Collapse
|
120
|
Fan W, Yung B, Huang P, Chen X. Nanotechnology for Multimodal Synergistic Cancer Therapy. Chem Rev 2017; 117:13566-13638. [DOI: 10.1021/acs.chemrev.7b00258] [Citation(s) in RCA: 1221] [Impact Index Per Article: 152.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Wenpei Fan
- Guangdong
Key Laboratory for Biomedical Measurements and Ultrasound Imaging,
School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
- Key
Laboratory of Optoelectronic Devices and Systems of Ministry of Education
and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Bryant Yung
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Peng Huang
- Guangdong
Key Laboratory for Biomedical Measurements and Ultrasound Imaging,
School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Xiaoyuan Chen
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
121
|
Bauer KN, Tee HT, Velencoso MM, Wurm FR. Main-chain poly(phosphoester)s: History, syntheses, degradation, bio-and flame-retardant applications. Prog Polym Sci 2017. [DOI: 10.1016/j.progpolymsci.2017.05.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
122
|
Wu X, Wang S, Li M, Wang A, Zhou Y, Li P, Wang Y. Nanocarriers for TRAIL delivery: driving TRAIL back on track for cancer therapy. NANOSCALE 2017; 9:13879-13904. [PMID: 28914952 DOI: 10.1039/c7nr04959e] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Since its initial identification, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has been shown to be capable of selectively inducing apoptosis in cancer cells. However, translation of the encouraging preclinical studies of this cytokine into the clinic has been restricted by its extremely short half-life, the presence of resistant cancer cell populations, and its inefficient in vivo delivery. Recently, there has been exceptional progress in developing novel formulations to increase the circulatory half-life of TRAIL and new combinations to treat cancers that are resistant to TRAIL. In particular, TRAIL-based nanotherapies offer the potential to improve the stability of TRAIL and prolong its half-life in plasma, to specifically deliver TRAIL to a particular target site, and to overcome resistance to TRAIL. The aim of this review is to provide an overview of the state-of-the art drug delivery systems that are currently being tested or developed to improve the biological attributes of TRAIL-based therapies.
Collapse
Affiliation(s)
- Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | | | | | | | | | | | | |
Collapse
|
123
|
Wang C, Du L, Zhou J, Meng L, Cheng Q, Wang C, Wang X, Zhao D, Huang Y, Zheng S, Cao H, Zhang J, Deng L, Liang Z, Dong A. Elaboration on the Distribution of Hydrophobic Segments in the Chains of Amphiphilic Cationic Polymers for Small Interfering RNA Delivery. ACS APPLIED MATERIALS & INTERFACES 2017; 9:32463-32474. [PMID: 28862422 DOI: 10.1021/acsami.7b07337] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Hydrophobization of cationic polymers, as an efficient strategy, had been widely developed in the structure of cationic polymer micelles to improve the delivery efficiency of nucleic acids. However, the distribution of hydrophobic segments in the polymer chains is rarely considered. Here, we have elaborated three types of hydrophobized polyethylene glycol (PEG)-blocked cationic polymers with different distributions of the hydrophobic segments in the polymer chains PEG-PAM-PDP (E-A-D), PEG-PDP-PAM (E-D-A), and PEG-P(AM/DP) (E-(A/D)), which were synthesized by reversible addition-fragmentation chain transfer polymerization of methoxy PEG, cationic monomer aminoethyl methacrylate, and pH-sensitive hydrophobic monomer 2-diisopropylaminoethyl methacrylate, respectively. In aqueous solution, all of the three copolymers, E-A-D, E-D-A, and E-(A/D), were able to spontaneously form nanosized micelles (100-150 nm) (ME-A-D, ME-D-A, and ME-(A/D)) and well-incorporated small interfering RNA (siRNA) into complex micelles (CMs). The effect of distributions of the hydrophobic segments on siRNA delivery had been evaluated in vitro and in vivo. Compared with ME-D-A and ME-(A/D), ME-A-D showed the best siRNA binding capacity to form stable ME-A-D/siRNA CMs less than 100 nm, mediated the best gene-silencing efficiency and inhibition effect of tumor cell growth in vitro, and showed better liver gene-silencing effect in vivo. In the case of ME-(A/D) with a random distribution of cationic and hydrophobic segments, a gene-silencing efficiency higher than Lipo2000 but lesser than ME-A-D and ME-D-A was obtained. As the mole ratio of positive and negative charges increased, ME-D-A/siRNA and ME-A-D/siRNA showed similar performances in size, zeta potential, cell uptake, and gene silencing, but ME-(A/D)/siRNA showed reversed performances. In addition, ME-A-D as the best siRNA carrier was evaluated in the tumor tissue in the xenograft murine model and showed good anticancer capacity. Obviously, the distribution of the hydrophobic segments in the amphiphilic cationic polymer chains should be seriously considered in the design of siRNA vectors.
Collapse
Affiliation(s)
- Changrong Wang
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , Tianjin 300072, China
| | | | - Junhui Zhou
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
| | | | | | - Chun Wang
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
| | | | | | - Yuanyu Huang
- Advanced Research Institute for Multidisciplinary Science, Beijing Institute of Technology , Beijing 100081, China
| | | | | | - Jianhua Zhang
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
| | - Liandong Deng
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
| | - Zicai Liang
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , Tianjin 300072, China
| | - Anjie Dong
- Department of Polymer Science and Technology, School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University , Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , Tianjin 300072, China
| |
Collapse
|
124
|
Iram S, Zahera M, Khan S, Khan I, Syed A, Ansary AA, Ameen F, Shair OHM, Khan MS. Gold nanoconjugates reinforce the potency of conjugated cisplatin and doxorubicin. Colloids Surf B Biointerfaces 2017; 160:254-264. [PMID: 28942160 DOI: 10.1016/j.colsurfb.2017.09.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 08/21/2017] [Accepted: 09/06/2017] [Indexed: 12/22/2022]
Abstract
Osteosarcoma or osteogenic sarcoma is the most common and prevalent cancerous tumor of bone and occurs especially in children and teens. Recent treatment strategy includes a combination of both chemotherapy and surgeries. Although, the use of single drug-based chemotherapy treatment remains unsatisfactory. Therefore, combinatorial therapy has emerged as a potential strategy for treatment with limited side- effects. Here, we evaluated the combinatorial anticancerous effect of cisplatin (CIS) and doxorubicin (DOX) bioconjugated bromelain encapsulated gold nanoparticles (B-AuNPs conjugated CIS and DOX) in the treatment of osteosarcoma. The synthesized B-AuNPs conjugated CIS and DOX were characterized by various characterization techniques like UV-vis spectroscopy, TEM, DLS and zeta potential to ensure the synthesis, size, shape, size distribution and stability. Drug loading efficiency bioconjugation of CIS and DOX was ensured by UV-vis spectroscopy. Bioconjugation of CIS and DOX was further confirmed using UV-vis spectroscopy, TEM, DLS, Zeta potential and FT-IR analysis. The combinatorial effect of CIS and DOX in B-AuNPs conjugated CIS and DOX showed highly improved potency against MG-63 and Saos-2 cells at a very low concentration where primary osteoblasts didn't show any cytotoxic effect. The apoptotic effect of B-AuNPs conjugated CIS and DOX on osteosarcoma and primary osteoblasts cells were analyzed by increased permeability of the cell membrane, condensed chromatin and deep blue fluorescent condensed nucleus. The results clearly showed that B-AuNPs conjugated CIS and DOX significantly improved the potency of both the chemotherapeutic drugs by delivering them specifically into the nucleus of cancer cells through caveolae-dependent endocytosis. Thus, the greater inhibitory effect of combinatorial drugs (B-AuNPs conjugated CIS and DOX) over single drug based chemotherapy would be of great advantage during osteosarcoma treatment.
Collapse
Affiliation(s)
- Sana Iram
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Manaal Zahera
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Salman Khan
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Imran Khan
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Abu Ayoobul Ansary
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune, 411008, India
| | - Fuad Ameen
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Omar H M Shair
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohd Sajid Khan
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India.
| |
Collapse
|
125
|
Abstract
BACKGROUND Nonhealing bone defects represent an immense biomedical burden. Despite recent advances in protein-based bone regeneration, safety concerns over bone morphogenetic protein-2 have prompted the search for alternative factors. Previously, the authors examined the additive/synergistic effects of hedgehog and Nel-like protein-1 (NELL-1) on the osteogenic differentiation of mesenchymal stem cells in vitro. In this study, the authors sought to leverage their previous findings by applying the combination of Smoothened agonist (SAG), hedgehog signal activator, and NELL-1 to an in vivo critical-size bone defect model. METHODS A 4-mm parietal bone defect was created in mixed-gender CD-1 mice. Treatment groups included control (n = 6), SAG (n = 7), NELL-1 (n = 7), and SAG plus NELL-1 (n = 7). A custom fabricated poly(lactic-co-glycolic acid) disk with hydroxyapatite coating was used as an osteoinductive scaffold. RESULTS Results at 4 and 8 weeks showed increased bone formation by micro-computed tomographic analyses with either stimulus alone (SAG or NELL-1), but significantly greater bone formation with both components combined (SAG plus NELL-1). This included greater bone healing scores and increased bone volume and bone thickness. Histologic analyses confirmed a significant increase in new bone formation with the combination therapy SAG plus NELL-1, accompanied by increased defect vascularization. CONCLUSIONS In summary, the authors' results suggest that combining the hedgehog signaling agonist SAG and NELL-1 has potential as a novel therapeutic strategy for the healing of critical-size bone defects. Future directions will include optimization of dosage and delivery strategy for an SAG and NELL-1 combination product.
Collapse
|
126
|
Wang M, Wang J, Li B, Meng L, Tian Z. Recent advances in mechanism-based chemotherapy drug-siRNA pairs in co-delivery systems for cancer: A review. Colloids Surf B Biointerfaces 2017; 157:297-308. [DOI: 10.1016/j.colsurfb.2017.06.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022]
|
127
|
Lin L, Liang X, Xu Y, Yang Y, Li X, Dai Z. Doxorubicin and Indocyanine Green Loaded Hybrid Bicelles for Fluorescence Imaging Guided Synergetic Chemo/Photothermal Therapy. Bioconjug Chem 2017; 28:2410-2419. [DOI: 10.1021/acs.bioconjchem.7b00407] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Li Lin
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Xiaolong Liang
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Yunxue Xu
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Yongbo Yang
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Xiaoda Li
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Zhifei Dai
- Department of Biomedical
Engineering, College of Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
128
|
Zhang R, Gao S, Wang Z, Han D, Liu L, Ma Q, Tan W, Tian J, Chen X. Multifunctional Molecular Beacon Micelles for Intracellular mRNA Imaging and Synergistic Therapy in Multidrug-Resistant Cancer Cells. ADVANCED FUNCTIONAL MATERIALS 2017; 27:1701027. [PMID: 29056886 PMCID: PMC5646829 DOI: 10.1002/adfm.201701027] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Multidrug resistance (MDR) resulting from overexpression of P-glycoprotein (Pgp) transporters increases the drug efflux and thereby limits the chemotherapeutic efficacy. It is desirable to administer both an MDR1 gene silencer and a chemotherapeutic agent in a sequential way to generate a synergistic therapeutic effect in multidrug-resistant cancer cells. Herein, we rationally designed an anti-MDR1 molecular beacon (MB)-based micelle (a-MBM) nanosystem, which is composed of a diacyllipid core densely packed with an MB corona. One of Pgp-transportable agents, doxorubicin (DOX), was encapsulated in the hydrophobic core of the micelle and in the stem sequence of MB. The a-MBM-DOX nanosystem showed an efficient self-delivery, enhanced enzymatic stability, excellent target selectivity, and high drug-loading capacity. With its relatively high enzymatic stability, a-MBM-DOX initially facilitated intracellular MDR1 mRNA imaging to distinguish multidrug-resistant and non-multidrug-resistant cells and subsequently downregulated the MDR1 gene expression owing to an antisense effect. After that, the MB corona was degraded, destroying the micellar nanostructure and releasing DOX, which resulted in a high accumulation of DOX in OVCAR8/ADR cells and a high chemotherapeutic efficacy owing to successful restoration of drug sensitivity. This micelle approach has the potential for both visualizing MDR1 mRNA and overcoming MDR in a sequential and synergistic way.
Collapse
Affiliation(s)
- Ruili Zhang
- China-Japan Union Hospital, Jilin University, Changchun, Jilin, 130033 China. Engineering Research Center of Molecular-imaging and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China. Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, 20892 USA
| | - Shi Gao
- China-Japan Union Hospital, Jilin University, Changchun, Jilin, 130033 China
| | - Zhongliang Wang
- Engineering Research Center of Molecular-imaging and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Da Han
- Center for Research at Bio/Nano Interface, University of Florida, Gainesville, FL 32611 USA
| | - Lin Liu
- China-Japan Union Hospital, Jilin University, Changchun, Jilin, 130033 China
| | - Qingjie Ma
- China-Japan Union Hospital, Jilin University, Changchun, Jilin, 130033 China
| | - Weihong Tan
- Center for Research at Bio/Nano Interface, University of Florida, Gainesville, FL 32611 USA
| | - Jie Tian
- Engineering Research Center of Molecular-imaging and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, 20892 USA
| |
Collapse
|
129
|
Zhang J, Li J, Shi Z, Yang Y, Xie X, Lee SM, Wang Y, Leong KW, Chen M. pH-sensitive polymeric nanoparticles for co-delivery of doxorubicin and curcumin to treat cancer via enhanced pro-apoptotic and anti-angiogenic activities. Acta Biomater 2017; 58:349-364. [PMID: 28455219 DOI: 10.1016/j.actbio.2017.04.029] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/22/2017] [Accepted: 04/24/2017] [Indexed: 01/24/2023]
Abstract
Co-delivery of multiple drugs with complementary anticancer mechanisms by nano-carriers offers an effective strategy to treat cancer. The combination of drugs with pro-apoptotic and anti-angiogenic activities is potentially effective in treating human hepatocellular carcinoma (HCC). Herein, we developed a co-delivery system for doxorubicin (Dox), a pro-apoptotic drug, and curcumin (Cur), a potent drug for antiangiogenesis, in pH-sensitive nanoparticles (NPs) constituted with amphiphilic poly(β-amino ester) copolymer. Dox & Cur co-loaded NPs ((D+C)/NPs) were prepared with optimized drug ratio, showing low polydispersity, high encapsulation efficiency, and enhanced release in the acidic environment of cancer cells. Furthermore, enhanced cellular internalization of cargoes delivered from (D+C)/NPs were observed in human liver cancer SMMC 7721 cells and human umbilical vein endothelial cells (HUVECs) compared to the use of free drugs. The (D+C)/NPs induced a high rate of apoptosis in SMMC 7721 cells through decreased mitochondrial membrane potential. Additionally, (D+C)/NPs exhibited stronger anti-angiogenic effects including inhibition of HUVEC proliferation, migration, invasion, and tube formation mediated VEGF pathway modulation in vitro and in vivo. Taken together, encapsulation of the pro-apoptotic drug Dox and antiangiogenic agent Cur in pH-sensitive NPs provides a promising strategy to effectively inhibit HCC progression in a synergistic manner. STATEMENT OF SIGNIFICANCE The combination of multiple drugs has been demonstrated to be more effective than single treatment. However, the different physicochemical and pharmacokinetic profiles of each drug render optimal delivery challenging. In view of the great delivery advantage of nanocarriers to unify the multiple drugs in vivo, stimulus-responsive nano-carriers are more crucial to increase efficacy and reduce toxicity from off-target exposure. Therefore, herein the pH-sensitive nanoparticles, composed by d-α-tocopheryl polyethylene glycol 1000-block-poly (β-amino ester) (TPGS-PAE) polymers, have been fabricated for doxorubicin (Dox) and curcumin (Cur) co-delivery, which exhibited diverse anticancer approaches, i.e. pro-apoptosis and antiangiogenesis. The precise intracellular target site and effective drug combination concentration result in the enhanced antitumor efficiency and the reduced systematic toxicity of Dox. The co-encapsulation of the pro-apoptotic drug and antiangiogenic agent in pH-sensitive NPs provides a promising strategy to effectively inhibit malignant neoplasm progression in a synergistic manner.
Collapse
|
130
|
Engineered polymeric nanoparticles to guide the cellular internalization and trafficking of small interfering ribonucleic acids. J Control Release 2017; 259:3-15. [DOI: 10.1016/j.jconrel.2017.02.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 12/29/2022]
|
131
|
Lee SH, Lee JY, Kim JS, Park TG, Mok H. Amphiphilic siRNA Conjugates for Co-Delivery of Nucleic Acids and Hydrophobic Drugs. Bioconjug Chem 2017; 28:2051-2061. [DOI: 10.1021/acs.bioconjchem.7b00222] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Soo Hyeon Lee
- Department
of Chemistry and Applied Biosciences, Institute of Pharmaceutical
Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), Vladimir-Prelog-Weg, Zurich 8093, Switzerland
| | - Jeong Yu Lee
- Basic Research & Innovation Division, AmorePacific Corporation R&D Unit, Yongin 446-729, Republic of Korea
| | - Jee Seon Kim
- National Institute of Biomedical Imaging and Bioengineering, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Tae Gwan Park
- Department
of Biological Science, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Hyejung Mok
- Department
of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| |
Collapse
|
132
|
Jiang Y, Sprouse D, Laaser JE, Dhande Y, Reineke TM, Lodge TP. Complexation of Linear DNA and Poly(styrenesulfonate) with Cationic Copolymer Micelles: Effect of Polyanion Flexibility. J Phys Chem B 2017; 121:6708-6720. [DOI: 10.1021/acs.jpcb.7b03732] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Yaming Jiang
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Ave. SE, Minneapolis, Minnesota 55455, United States
| | - Dustin Sprouse
- Department
of Chemistry, University of Minnesota, 207 Pleasant St. SE, Minneapolis, Minnesota 55455, United States
| | - Jennifer E. Laaser
- Department
of Chemistry, University of Minnesota, 207 Pleasant St. SE, Minneapolis, Minnesota 55455, United States
| | - Yogesh Dhande
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Ave. SE, Minneapolis, Minnesota 55455, United States
| | - Theresa M. Reineke
- Department
of Chemistry, University of Minnesota, 207 Pleasant St. SE, Minneapolis, Minnesota 55455, United States
| | - Timothy P. Lodge
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Ave. SE, Minneapolis, Minnesota 55455, United States
- Department
of Chemistry, University of Minnesota, 207 Pleasant St. SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
133
|
Chen X, Zhang Y, Tang C, Tian C, Sun Q, Su Z, Xue L, Yin Y, Ju C, Zhang C. Co-delivery of paclitaxel and anti-survivin siRNA via redox-sensitive oligopeptide liposomes for the synergistic treatment of breast cancer and metastasis. Int J Pharm 2017. [PMID: 28642204 DOI: 10.1016/j.ijpharm.2017.06.071] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The overexpression of survivin in breast cancer cells is an important factor of paclitaxel (PTX) resistance in breast cancer. To overcome PTX resistance and improve the antitumor effect of PTX, we developed a novel liposome-based nanosystem (PTX/siRNA/SS-L), composed of a redox-sensitive cationic oligopeptide lipid (LHSSG2C14) with a proton sponge effect, natural soybean phosphatidylcholine (SPC), and cholesterol for co-delivery of PTX and anti-survivin siRNA, which could specifically downregulate survivin overexpression. PTX/siRNA/SS-L exhibited high encapsulation efficiency and rapid redox-responsive release of both PTX and siRNA. Moreover, in vitro studies on the 4T1 breast cancer cells revealed that PTX/siRNA/SS-L offered significant advantages over other experimental groups, such as higher cellular uptake, successful endolysosomal escape, reduced survivin expression, the lowest cell viability and wound healing rate, as well as the highest apoptosis rate. In particular, in vivo evaluation of 4T1 tumor-bearing mice showed that PTX/siRNA/SS-L had lower toxicity and induced a synergistic inhibitory effect on tumor growth and pulmonary metastasis. Collectively, the collaboration of anti-survivin siRNA and PTX via redox-sensitive oligopeptide liposomes provides a promising strategy for the treatment of breast cancer and metastasis.
Collapse
Affiliation(s)
- Xinyan Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China; Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yidi Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Chunming Tang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Chunli Tian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Qiong Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Zhigui Su
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Yifan Yin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China
| | - Caoyun Ju
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China.
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210046, China.
| |
Collapse
|
134
|
Wang Q, Liu S, Yang F, Gan L, Yang X, Yang Y. Magnetic alginate microspheres detected by MRI fabricated using microfluidic technique and release behavior of encapsulated dual drugs. Int J Nanomedicine 2017; 12:4335-4347. [PMID: 28652736 PMCID: PMC5473605 DOI: 10.2147/ijn.s131249] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Alginate microspheres loaded with superparamagnetic iron oxide nanoparticles (SPIO NPs) have been fabricated by a T-junction microfluidic device combined with an external ionic crosslinking. The obtained microspheres possess excellent visuality under magnetic resonance due to the presence of only 0.6 mg/mL SPIO NPs. The microspheres also show uniform size with narrow distribution and regular spherical shape characterized by optic microscope and environmental scanning electron microscope. Furthermore, dual drugs (5-fluorouracil and doxorubicin hydrochloride) have been loaded within the microspheres. The release behavior of dual drugs from the microspheres show typical sustained release profiles. As a novel embolic agent, such microspheres in blood vessels can be tracked by magnetic resonance scanner. Thus, the integration of embolotherapy, chemotherapy, and postoperative diagnosis can be realized.
Collapse
Affiliation(s)
- Qin Wang
- National Engineering Research Center for Nanomedicine, School of Chemistry and Chemical Engineering
| | - Shanshan Liu
- National Engineering Research Center for Nanomedicine, School of Chemistry and Chemical Engineering
| | - Fan Yang
- Department of Radiology, Union Hospital, Tongji Medical College
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yajiang Yang
- National Engineering Research Center for Nanomedicine, School of Chemistry and Chemical Engineering
| |
Collapse
|
135
|
Chen MM, Liu YY, Su GH, Song FF, Liu Y, Zhang QQ. NIR responsive liposomal system for rapid release of drugs in cancer therapy. Int J Nanomedicine 2017; 12:4225-4239. [PMID: 28652729 PMCID: PMC5473596 DOI: 10.2147/ijn.s130861] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
To design a rapid release liposomal system for cancer therapy, a NIR responsive bubble-generating thermosensitive liposome (BTSL) system combined with photothermal agent (Cypate), doxorubicin (DOX), and NH4HCO3 was developed. Cypate/DOX-BTSL exhibited a good aqueous stability, photostability, and photothermal effect. In vitro release suggested that the amounts of DOX released from BTSL were obviously higher than that of (NH4)2SO4 liposomes at 42°C. After NIR irradiation, the hyperthermic temperature induced by Cypate led to the decomposition of NH4HCO3 and the generation of a large number of CO2 bubbles, triggering a rapid release of drugs. Confocal laser scanning microscope and acridine orange staining indicated that Cypate/DOX-BTSL upon irradiation could facilitate to disrupt the lysosomal membranes and realize endolysosomal escape into cytosol, improving the intracellular uptake of DOX clearly. MTT and trypan blue staining implied that the cell damage of Cypate/DOX-BTSL with NIR irradiation was more severe than that in the groups without irradiation. In vivo results indicated that Cypate/DOX-BTSL with irradiation could dramatically increase the accumulation of DOX in tumor, inhibit tumor growth, and reduce systemic side effects of DOX. These data demonstrated that Cypate/DOX-BTSL has the potential to be used as a NIR responsive liposomal system for a rapid release of drugs in thermochemotherapy.
Collapse
Affiliation(s)
- Ming-Mao Chen
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou
| | - Yuan-Yuan Liu
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou
| | - Guang-Hao Su
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou
| | - Fei-Fei Song
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou
| | - Yan Liu
- State Key Lab of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou
| | - Qi-Qing Zhang
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University, Fuzhou
- Key Laboratory of Biomedical Material of Tianjin, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, People’s Republic of China
| |
Collapse
|
136
|
Pina MF, Lau W, Scherer K, Parhizkar M, Edirisinghe M, Craig D. The generation of compartmentalized nanoparticles containing siRNA and cisplatin using a multi-needle electrohydrodynamic strategy. NANOSCALE 2017; 9:5975-5985. [PMID: 28440835 DOI: 10.1039/c7nr01002h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
This study outlines a novel manufacturing technique for the generation of compartmentalized trilayered nanoparticles loaded with an anti-cancer agent and siRNA as a platform for the combination treatment of cancers. More specifically, we describe the use of a multi-needle electrohydrodynamic approach to produce nanoparticles with high size specificity and scalable output, while allowing suitable environments for each therapeutic agent. The inner polylactic-glycolic-acid (PLGA) layer was loaded with cisplatin while the middle chitosan layer was loaded with siRNA. The corresponding polymeric solutions were characterized for their viscosity, surface tension and conductivity, while particle size was determined using dynamic light scattering. The internal structure was studied using transmission electron microscopy (TEM) and Structured Illumination Microscopy (SIM). The inclusion of cisplatin was studied using electron dispersive spectroscopy (EDS). We were able to generate nanoparticles of approximate size 130 nm with three distinct layers containing an outer protective PLGA layer, a middle layer of siRNA and an inner layer of cisplatin. These particles have the potential not only for uptake into tumors via the enhanced permeability and retention (EPR) effect but also the sequential release of the siRNA and chemotherapeutic agent, thereby providing a means of overcoming challenges of targeting and tumor drug resistance.
Collapse
Affiliation(s)
- Maria F Pina
- University College London School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Wai Lau
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | - Kathrin Scherer
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Maryam Parhizkar
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | - Mohan Edirisinghe
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | - Duncan Craig
- University College London School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
137
|
Yu M, Han S, Kou Z, Dai J, Liu J, Wei C, Li Y, Jiang L, Sun Y. Lipid nanoparticle-based co-delivery of epirubicin and BCL-2 siRNA for enhanced intracellular drug release and reversing multidrug resistance. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:323-332. [PMID: 28393563 DOI: 10.1080/21691401.2017.1307215] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
At present, combined therapy has become an effective strategy for the treatment of cancer. Co-delivery of the chemotherapeutic drugs and siRNA can more effectively inhibit tumor growth by nano drug delivery systems (NDDSs). Here, we prepared and evaluated a multifunctional envelope-type nano device (MEND). This MEND was a kind of composite lipid-nanoparticles possessing both the properties of liposomes and nanoparticles. In this study, an acid-cleavable ketal containing poly (β-amino ester) (KPAE) was used to bind siBCL-2 and the KPAE/siBCL-2 complexes were further coated by epirubicin (EPI) containing lipid to form EPI/siBCL-2 dual loaded lipid-nanoparticles. The results showed that the average size of EPI/siBCL-2-MEND was about 120 nm, and the average zeta potential was about 41 mV. The encapsulation efficiency (EE) of EPI and siBCL-2 was 86.13% and 97.07%, respectively. EPI/siBCL-2 dual loaded lipid-nanoparticles showed enhanced inhibition efficiency than individual EPI-loaded liposomes on HepG2 cells by MTT assay. Moreover, western blot experiment indicated co-delivery of EPI/siBCL-2 can significantly down-regulate the expression of P-glycoprotein (P-gp), while free EPI and EPI-loaded liposomes up-regulated it. Therefore, the strategy of co-delivering EPI and siBCL-2 simultaneously by lipid-nanoparticles showed promising potential in reversing multidrug resistance of tumor cells.
Collapse
Affiliation(s)
- Miao Yu
- a School of Pharmacy, Qingdao University , Qingdao , China
| | - Shangcong Han
- a School of Pharmacy, Qingdao University , Qingdao , China
| | - Zhongai Kou
- b Department of Neurology , Shengli Hospital , Dongying , China
| | - Jialing Dai
- a School of Pharmacy, Qingdao University , Qingdao , China
| | - Jiao Liu
- a School of Pharmacy, Qingdao University , Qingdao , China
| | - Chen Wei
- a School of Pharmacy, Qingdao University , Qingdao , China
| | - Yitong Li
- a School of Pharmacy, Qingdao University , Qingdao , China
| | - Lutao Jiang
- a School of Pharmacy, Qingdao University , Qingdao , China
| | - Yong Sun
- a School of Pharmacy, Qingdao University , Qingdao , China
| |
Collapse
|
138
|
Tatiparti K, Sau S, Kashaw SK, Iyer AK. siRNA Delivery Strategies: A Comprehensive Review of Recent Developments. NANOMATERIALS (BASEL, SWITZERLAND) 2017; 7:E77. [PMID: 28379201 PMCID: PMC5408169 DOI: 10.3390/nano7040077] [Citation(s) in RCA: 282] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/07/2017] [Accepted: 03/31/2017] [Indexed: 01/01/2023]
Abstract
siRNA is a promising therapeutic solution to address gene overexpression or mutations as a post-transcriptional gene regulation process for several pathological conditions such as viral infections, cancer, genetic disorders, and autoimmune disorders like arthritis. This therapeutic method is currently being actively pursued in cancer therapy because siRNA has been found to suppress the oncogenes and address mutations in tumor suppressor genes and elucidate the key molecules in cellular pathways in cancer. It is also effective in personalized gene therapy for several diseases due to its specificity, adaptability, and broad targeting capability. However, naked siRNA is unstable in the bloodstream and cannot efficiently cross cell membranes besides being immunogenic. Therefore, careful design of the delivery systems is essential to fully utilize the potential of this therapeutic solution. This review presents a comprehensive update on the challenges of siRNA delivery and the current strategies used to develop nanoparticulate delivery systems.
Collapse
Affiliation(s)
- Katyayani Tatiparti
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| | - Samaresh Sau
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| | - Sushil Kumar Kashaw
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar 470003, India.
| | - Arun K Iyer
- Use-Inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
139
|
Wang Q, Jiang H, Li Y, Chen W, Li H, Peng K, Zhang Z, Sun X. Targeting NF-kB signaling with polymeric hybrid micelles that co-deliver siRNA and dexamethasone for arthritis therapy. Biomaterials 2017; 122:10-22. [DOI: 10.1016/j.biomaterials.2017.01.008] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/04/2017] [Accepted: 01/07/2017] [Indexed: 12/17/2022]
|
140
|
Wang H, Li Y, Zhang M, Wu D, Shen Y, Tang G, Ping Y. Redox-Activatable ATP-Depleting Micelles with Dual Modulation Characteristics for Multidrug-Resistant Cancer Therapy. Adv Healthc Mater 2017; 6. [PMID: 28152267 DOI: 10.1002/adhm.201601293] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/08/2017] [Indexed: 11/12/2022]
Abstract
A fast adenosine triphosphate (ATP)-depleting micellar system that is activated by intracellular redox for the codelivery of anticancer drug paclitaxel (PTX) and small interference RNA (siRNA) targeting polo-like kinase1 (PLK1) is developed to address the key challenges of multidrug-resistant (MDR) cancer therapy. The ATP-depleting micelle is self-assembled from a redox-responsive amphiphilic polymer (termed as bPEG-SS-P123-PEI (PSPP)) that is composed of biocompatible branched polyethylene glycol (PEG) with 8 arms (bPEG), ATP-depleting Pluronic P123 (P123), and cationic low molecular weight polyethylenimine (PEI) blocks. Upon critical micelle concentration, the PSPP unimer self-assembles into a well-ordered multilayered nanostructure and is able to load PTX and siRNA targeting PLK1. The cleavage of disulfide linkages at intracellular glutathione-rich reduction milieu not only promotes PTX and siRNA release, but also activates the fast ATP-depletion action that is critical in preventing intracellular PTX efflux by multidrug-resistant cancer cells. The combination of ATP depletion and siRNA inhibition by PSPP micelles is found to provide dual modulations for resensitizing multidrug-resistant cancer cells for PTX treatment. As a result, the codelivery of PTX and PLK1 siRNA exerts a stronger combinational effect against tumor growth in MDR tumor models in vivo. The development of fast ATP-depleting nanomicelle represents an original delivery strategy for the distinctive dual modulation of cancer MDR with spatial and temporal control.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Animals
- Cell Cycle Proteins/antagonists & inhibitors
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Line, Tumor
- Drug Delivery Systems/methods
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Multiple/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- HEK293 Cells
- Humans
- Mice
- Mice, Inbred BALB C
- Micelles
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Oxidation-Reduction
- Paclitaxel/pharmacology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/pharmacology
- Polo-Like Kinase 1
Collapse
Affiliation(s)
- Hebin Wang
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
- College of life Sciences, Tarim University, Alar, 843300, China
| | - Yang Li
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Miaozun Zhang
- Department of General Surgery, Ningbo Li Huili Hospital, Medical School of Ningbo University, Ningbo, 315040, China
| | - Di Wu
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Youqing Shen
- Center for Bionanoengineering and State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Guping Tang
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Yuan Ping
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| |
Collapse
|
141
|
Yousefpour Marzbali M, Yari Khosroushahi A. Polymeric micelles as mighty nanocarriers for cancer gene therapy: a review. Cancer Chemother Pharmacol 2017; 79:637-649. [DOI: 10.1007/s00280-017-3273-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 03/02/2017] [Indexed: 12/11/2022]
|
142
|
He YJ, Xing L, Cui PF, Zhang JL, Qiao JB, Luo CQ, Jiang G, Jiang HL. Branched polymeric prodrug/programmed cell death 4 complexes for combinational cancer therapy. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2017. [DOI: 10.1007/s40005-016-0301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
143
|
Elzeny H, Zhang F, Ali EN, Fathi HA, Zhang S, Li R, El-Mokhtar MA, Hamad MA, Wooley KL, Elsabahy M. Polyphosphoester nanoparticles as biodegradable platform for delivery of multiple drugs and siRNA. Drug Des Devel Ther 2017; 11:483-496. [PMID: 28260861 PMCID: PMC5327906 DOI: 10.2147/dddt.s128503] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Delivery of multiple therapeutics and/or diagnostic agents to diseased tissues is challenging and necessitates the development of multifunctional platforms. Among the various strategies for design of multifunctional nanocarriers, biodegradable polyphosphoester (PPE) polymers have been recently synthesized via a rapid and simple synthetic strategy. In addition, the chemical structure of the polymer could be tuned to form nanoparticles with varying surface chemistries and charges, which have shown exceptional safety and biocompatibility as compared to several commercial agents. The purpose of this study was to exploit a mixture of PPE nanoparticles of cationic and neutral surface charges for multiple delivery of anticancer drugs (ie, sorafenib and paclitaxel) and nucleic acids (ie, siRNA). Cationic PPE polymers could efficiently complex siRNA, and the stability of the nanoparticles could be maintained in physiological solutions and upon freeze-drying and were able to deliver siRNA in vivo when injected intravenously in mice. Commercially available cationic polyethylenimine polymer had LD50 of ca. 61.7 mg/kg in mice, whereas no animal died after injection of the cationic PPE polymer at a dose of >130 mg/kg. Neutral PPE nanoparticles were able to encapsulate two hydrophobic drugs, namely, sorafenib and paclitaxel, which are commonly used for the treatment of hepatocellular carcinoma. Mixing the neutral and cationic PPE nanoparticles did not result in any precipitation, and the size characteristics of both types of nanoparticles were maintained. Hence, PPE polymers might have potential for the delivery of multiple drugs and diagnostic agents to diseased tissues via simple synthesis of the individual polymers and assembly into nanoparticles that can host several drugs while being mixed in the same administration set, which is of importance for industrial and clinical development.
Collapse
Affiliation(s)
- Hadeel Elzeny
- Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut, Egypt
| | - Fuwu Zhang
- Departments of Chemistry, Chemical Engineering and Materials Science and Engineering, Texas A&M University, College Station, TX, USA
| | - Esraa N Ali
- Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut, Egypt
| | - Heba A Fathi
- Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut, Egypt
| | - Shiyi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Richen Li
- Departments of Chemistry, Chemical Engineering and Materials Science and Engineering, Texas A&M University, College Station, TX, USA
| | | | - Mostafa A Hamad
- Department of Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Karen L Wooley
- Departments of Chemistry, Chemical Engineering and Materials Science and Engineering, Texas A&M University, College Station, TX, USA
- Laboratory for Synthetic-Biologic Interactions, Department of Chemistry, Texas A&M University, College Station, TX, USA
| | - Mahmoud Elsabahy
- Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut, Egypt
- Laboratory for Synthetic-Biologic Interactions, Department of Chemistry, Texas A&M University, College Station, TX, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut
- Misr University for Science and Technology, 6th of October City, Egypt
| |
Collapse
|
144
|
A drug-delivery strategy for overcoming drug resistance in breast cancer through targeting of oncofetal fibronectin. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:713-722. [DOI: 10.1016/j.nano.2016.10.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/20/2022]
|
145
|
Recent advances in the design, development, and targeting mechanisms of polymeric micelles for delivery of siRNA in cancer therapy. Prog Polym Sci 2017. [DOI: 10.1016/j.progpolymsci.2016.09.008] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
146
|
Chen X, Yuan P, Liu Z, Bai Y, Zhou Y. Dual responsive hydrogels based on functionalized mesoporous silica nanoparticles as an injectable platform for tumor therapy and tissue regeneration. J Mater Chem B 2017; 5:5968-5973. [DOI: 10.1039/c7tb01225j] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A dual responsive hydrogel based on MSNs was fabricated to offer a strategy for sequential tumor therapy and tissue regeneration.
Collapse
Affiliation(s)
- Xin Chen
- School of Chemical Engineering and Technology
- Shanxi Key Laboratory of Energy Chemical Process Intensification
- Institute of Polymer Science in Chemical Engineering
- Xi'an Jiao Tong University
- Xi'an
| | - Pingyun Yuan
- School of Chemical Engineering and Technology
- Shanxi Key Laboratory of Energy Chemical Process Intensification
- Institute of Polymer Science in Chemical Engineering
- Xi'an Jiao Tong University
- Xi'an
| | - Zhongning Liu
- Department of Prosthodontics
- Peking University School and Hospital of Stomatology
- Beijing 100081
- China
| | - Yongkang Bai
- School of Chemical Engineering and Technology
- Shanxi Key Laboratory of Energy Chemical Process Intensification
- Institute of Polymer Science in Chemical Engineering
- Xi'an Jiao Tong University
- Xi'an
| | - Yongsheng Zhou
- Department of Prosthodontics
- Peking University School and Hospital of Stomatology
- Beijing 100081
- China
| |
Collapse
|
147
|
Li Y, Wang H, Wang K, Hu Q, Yao Q, Shen Y, Yu G, Tang G. Targeted Co-delivery of PTX and TR3 siRNA by PTP Peptide Modified Dendrimer for the Treatment of Pancreatic Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1602697. [PMID: 27762495 DOI: 10.1002/smll.201602697] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Indexed: 06/06/2023]
Abstract
A new type of tumor-targeted nanovehicle peptide-conjugated PSPG (PSPGP) is successfully synthesized for co-delivery of paclitaxel (PTX) and TR3 small interfering RNA (siRNA). In vitro and in vivo investigations demonstrate that the redox-responsive PSPGP exhibit enhanced endosomal escape and intracellular degradation, which facilitate PTX and TR3 siRNA release, effectively improving the antitumor efficacy.
Collapse
Affiliation(s)
- Yang Li
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Hebin Wang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
- College of Life Sciences, Tarim University, Alar, 843300, China
| | - Kai Wang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Qi Yao
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Youqing Shen
- Center for Bionanoengineering and State Key Laboratory for Chemical Engineering, Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Guocan Yu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Guping Tang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| |
Collapse
|
148
|
Nanocarrier-based co-delivery of small molecules and siRNA/miRNA for treatment of cancer. Ther Deliv 2016; 7:245-55. [PMID: 27010986 DOI: 10.4155/tde-2015-0003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aberrant gene expression can trigger several vital molecular events that not only result in carcinogenesis but also cause chemoresistance, metastasis and relapse. Gene-based therapies using siRNA/miRNA have been suggested as new treatment method to improve the current regimen. Although these agents can restore the normal molecular cascade thereby resensitizing the cancer cells, delivering a standard regimen (either subsequently or simultaneously) is necessary to achieve the therapeutic benefit. However, co-delivery using a single carrier could give an additional advantage of similar biodistribution profile of the loaded agents. While much research has been carried out in this field in recent years, challenges involved in designing combination formulations including efficient coloading, stability, appropriate biodistribution and target specificity have hampered their clinical translation. This article highlights current aspects of nano-carriers used for co-delivery of small molecules and genes to treat cancer.
Collapse
|
149
|
Yin T, Liu J, Zhao Z, Dong L, Cai H, Yin L, Zhou J, Huo M. Smart nanoparticles with a detachable outer shell for maximized synergistic antitumor efficacy of therapeutics with varying physicochemical properties. J Control Release 2016; 243:54-68. [DOI: 10.1016/j.jconrel.2016.09.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/29/2016] [Accepted: 09/30/2016] [Indexed: 01/07/2023]
|
150
|
Jeong H, Lee SH, Hwang Y, Yoo H, Jung H, Kim SH, Mok H. Multivalent Aptamer-RNA Conjugates for Simple and Efficient Delivery of Doxorubicin/siRNA into Multidrug-Resistant Cells. Macromol Biosci 2016; 17. [PMID: 27863037 DOI: 10.1002/mabi.201600343] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/07/2016] [Indexed: 01/08/2023]
Abstract
Multivalent aptamer-siRNA conjugates containing multiple mucin-1 aptamers and BCL2-specific siRNA are synthesized, and doxorubicin, an anthracycline anticancer drug, is loaded into these conjugates through intercalation with nucleic acids. These doxorubicin-incorporated multivalent aptamer-siRNA conjugates are transfected to mucin-1 overexpressing MCF-7 breast cancer cells and their multidrug-resistant cell lines. Doxorubicin-incorporated multivalent aptamer-siRNA conjugates exert promising anticancer effects, such as activation of caspase-3/7 and decrease of cell viability, on multidrug-resistant cancer cells because of their high intracellular uptake efficiency. Thus, this delivery system is an efficient tool for combination oncotherapy with chemotherapeutics and nucleic acid drugs to overcome multidrug resistance.
Collapse
Affiliation(s)
- Hyosook Jeong
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Soo Hyeon Lee
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology Zurich (ETHZ), 8093 Zurich, Switzerland
| | - Yeonju Hwang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Hyundong Yoo
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Heesun Jung
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sun Hwa Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 136-791, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| |
Collapse
|