1
|
Thapa B, Kc R, Uludağ H. TRAIL therapy and prospective developments for cancer treatment. J Control Release 2020; 326:335-349. [PMID: 32682900 DOI: 10.1016/j.jconrel.2020.07.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/01/2020] [Accepted: 07/11/2020] [Indexed: 12/22/2022]
Abstract
Tumor Necrosis Factor (TNF) Related Apoptosis-Inducing Ligand (TRAIL), an immune cytokine of TNF-family, has received much attention in late 1990s as a potential cancer therapeutics due to its selective ability to induce apoptosis in cancer cells. TRAIL binds to cell surface death receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5) and facilitates formation of death-inducing signaling complex (DISC), eventually activating the p53-independent apoptotic cascade. This unique mechanism makes the TRAIL a potential anticancer therapeutic especially for p53-mutated tumors. However, recombinant human TRAIL protein (rhTRAIL) and TRAIL-R agonist monoclonal antibodies (mAb) failed to exert robust anticancer activities due to inherent and/or acquired resistance, poor pharmacokinetics and weak potencies for apoptosis induction. To get TRAIL back on track as a cancer therapeutic, multiple strategies including protein modification, combinatorial approach and TRAIL gene therapy are being extensively explored. These strategies aim to enhance the half-life and bioavailability of TRAIL and synergize with TRAIL action ultimately sensitizing the resistant and non-responsive cells. We summarize emerging strategies for enhanced TRAIL therapy in this review and cover a wide range of recent technologies that will provide impetus to rejuvenate the TRAIL therapeutics in the clinical realm.
Collapse
Affiliation(s)
- Bindu Thapa
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| | - Remant Kc
- Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada.
| | - Hasan Uludağ
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada; Department of Biomedical Engineering, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
2
|
Down‐regulation of intracellular anti‐apoptotic proteins, particularly c‐FLIP by therapeutic agents; the novel view to overcome resistance to TRAIL. J Cell Physiol 2018; 233:6470-6485. [PMID: 29741767 DOI: 10.1002/jcp.26585] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/08/2018] [Indexed: 12/24/2022]
|
3
|
Liu Y, Kim YJ, Siriwon N, Rohrs JA, Yu Z, Wanga P. Combination drug delivery via multilamellar vesicles enables targeting of tumor cells and tumor vasculature. Biotechnol Bioeng 2018; 115:1403-1415. [PMID: 29457630 DOI: 10.1002/bit.26566] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/16/2018] [Accepted: 02/08/2018] [Indexed: 12/15/2022]
Abstract
Blood vessel development is critical for the continued growth and progression of solid tumors and, therefore, makes an attractive target for improving cancer therapy. Indeed, vascular-targeted therapies have been extensively explored but they have shown minimal efficacy as monotherapies. Combretastatin A4 (CA-4) is a tubulin-binding vascular disrupting agent that selectively targets the established tumor endothelium, causing rapid vascular beak down. Despite its potent anticancer potential, the drug has dose-limiting side effects, particularly in the form of cardiovascular toxicity. Furthermore, its poor aqueous solubility and the resulting limited bioavailability hinder its antitumor activity in the clinic. To improve the therapeutic efficacy of CA-4, we investigated its application as a combination therapy with doxorubicin (Dox) in a tumor vasculature targeted delivery vehicle: peptide-modified cross-linked multilamellar liposomal vesicles (cMLVs). In vitro cell culture studies showed that a tumor vasculature-targeting peptide, RIF7, could facilitate higher cellular uptake of drug-loaded cMLVs, and consequently enhance the antitumor efficacy in both drug resistant B16 mouse melanoma and human MDA-MB-231 breast cancer cells. In vivo, upon intravenous injection, targeted cMLVs could efficiently deliver both Dox and CA-4 to significantly slow tumor growth through the specific interaction of the targeting peptide with its receptor on the surface of tumor vasculature. This study demonstrates the potential of our novel targeted combination therapy delivery vehicle to improve the outcome of cancer treatment.
Collapse
Affiliation(s)
- Yarong Liu
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
| | - Yu J Kim
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California
| | - Natnaree Siriwon
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California
| | - Jennifer A Rohrs
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, Guangdong, China
| | - Pin Wanga
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California.,Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California.,Department of Biomedical Engineering, University of Southern California, Los Angeles, California
| |
Collapse
|
4
|
Xiao W, Xiong J, Zhang S, Xiong Y, Zhang H, Gao H. Influence of ligands property and particle size of gold nanoparticles on the protein adsorption and corresponding targeting ability. Int J Pharm 2018; 538:105-111. [DOI: 10.1016/j.ijpharm.2018.01.011] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/20/2017] [Accepted: 01/03/2018] [Indexed: 11/29/2022]
|
5
|
Kim TE, Hong S, Song K, Park SH, Shin YK. Sensitization of glycoengineered interferon-β1a-resistant cancer cells by cFLIP inhibition for enhanced anti-cancer therapy. Oncotarget 2017; 8:13957-13970. [PMID: 28086218 PMCID: PMC5355153 DOI: 10.18632/oncotarget.14573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/27/2016] [Indexed: 12/30/2022] Open
Abstract
In this study, we examined the molecular mechanism underlying the resistance of cancer cells to R27T, a glycoengineered version of recombinant human interferon (IFN)-β1a, and sought to overcome R27T resistance through combination therapy. R27T has been shown to induce anti-proliferation and apoptosis in human OVCAR-3 and MCF-7 cells, but not in HeLa cells. R27T treatment increased caspase-8 activity and the consequent cleavage of caspase-8 and -3 in R27T-sensitive OVCAR-3 cells, but not in R27T-resistant HeLa cells. Conversely, R27T increased the expression of cellular FLICE-like inhibitory protein (cFLIP) in HeLa cells, but not in OVCAR-3 cells. The sensitization of HeLa cells with cFLIP small interfering RNA or 4,5,6,7-tetrabromobenzotriazole (TBB, an inhibitor of casein kinase-2) facilitated R27T-induced caspase activation, and consequently apoptosis. In OVCAR-3-xenografted mice, intraperitoneal administration of R27T showed 2.1-fold higher anti-tumor efficacy than did the control vehicle. The combined administration of R27T and TBB showed the greatest anti-tumor effect in HeLa tumor-bearing mice, reducing the relative tumor volume by 35.7% compared to that in R27T-treated mice. Taken together, our results suggest that R27T has potential as an anti-cancer drug, and combination therapy with cFLIP inhibitors may be an effective strategy for overcoming R27T resistance.
Collapse
Affiliation(s)
- Tae-Eun Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungyoul Hong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyoung Song
- Abion Inc., R&D Center, Seoul 08394, Republic of Korea
| | - Sang-Ho Park
- Abion Inc., R&D Center, Seoul 08394, Republic of Korea.,GE Healthcare Korea, R&D Center, Incheon 21988, Republic of Korea
| | - Young Kee Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
6
|
MiR-126 reverses drug resistance to TRAIL through inhibiting the expression of c-FLIP in cervical cancer. Gene 2017; 627:420-427. [DOI: 10.1016/j.gene.2017.06.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/19/2017] [Accepted: 06/28/2017] [Indexed: 01/20/2023]
|
7
|
Tam C, Wong JH, Cheung RCF, Zuo T, Ng TB. Therapeutic potentials of short interfering RNAs. Appl Microbiol Biotechnol 2017; 101:7091-7111. [PMID: 28791440 DOI: 10.1007/s00253-017-8433-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/19/2017] [Indexed: 01/10/2023]
Abstract
Short interfering RNA (siRNA) is one of the members of the family of RNA interference (RNAi). Coupled with the RNA-induced silencing complex (RISC), siRNA is able to trigger the cleavage of target RNAs which serve as a defensive system against pathogens. Meanwhile, siRNA in gene silencing opens a new avenue for the treatment of various diseases. SiRNA can effectively inhibit viral infection and replication and suppress tumorigenesis and various inflammation-associated diseases and cardiovascular diseases by inactivation of viral genes and downregulation of oncogene expression. Recently, endogenous siRNAs (endo-siRNAs) were discovered in the reproductive cells of animals which may be associated with regulation of cell division. Structural modification of siRNA enhances the delivery, specificity and efficacy and bioavailability to the target cells. There are at least five categories of siRNA delivery systems including viral vectors, lipid-based nanoparticles, peptide-based nanoparticles, polymer-based nanoparticles and inorganic small molecules like metal ions, silica and carbon. Sufficient preclinical and clinical studies supported that siRNA may be a potential medicine for targeted therapy of various diseases in the near future.
Collapse
Affiliation(s)
- Chit Tam
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Sha Tin, New Territories, Hong Kong, China.
| | - Jack Ho Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Sha Tin, New Territories, Hong Kong, China
| | - Randy Chi Fai Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Sha Tin, New Territories, Hong Kong, China
| | - Tao Zuo
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong, China
| | - Tzi Bun Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Sha Tin, New Territories, Hong Kong, China.
| |
Collapse
|
8
|
Cao C, Luo X, Ji X, Wang Y, Zhang Y, Zhang P, Zhong L. Osteopontin regulates the proliferation of rat aortic smooth muscle cells in response to gingipains treatment. Mol Cell Probes 2017; 33:51-56. [PMID: 28302392 DOI: 10.1016/j.mcp.2017.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 03/11/2017] [Accepted: 03/11/2017] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The present study aimed to explore the possible effects of osteopontin (OPN) in the proliferation of rat aortic smooth muscle cells (RASMCs) stimulated by gingipains. METHODS The proliferation of RASMCs in response to active gingipains treatment was evaluated by CCK-8 assay. OPN siRNA was designed, constructed and transfected into RASMCs at different concentrations. The cell cycle of RASMCs was analyzed by flow cytometry. OPN, α-SMA and calponin expression were examined by real-time PCR and western blot analysis. RESULTS Gingipains promoted the proliferation of RASMCs and OPN expression. With siRNA-mediated OPN expression knockdown, the cell cycle of RASMCs was blocked in the G0/G1 phase. Furthermore, the expression of specific differentiation markers, α-SMA and calponin, also decreased. CONCLUSIONS These results demonstrate that OPN has an impact on the proliferation and differentiation of RASMCs stimulated by gingipains.
Collapse
Affiliation(s)
- Chong Cao
- Department of Periodontology, Caochong Dental Clinic, Urumqi 830054, China
| | - Xin Luo
- Department of Pharmacology, The Basic Medical Sciences College of Xinjiang Medical University, Urumqi 830054, China
| | - Xiaowei Ji
- Department of Periodontology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Yao Wang
- Department of Stomatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
| | - Yuan Zhang
- Medical College of Hangzhou Normal University, Hangzhou 311121, China; Department of Stomatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
| | - Pengtao Zhang
- Medical College of Hangzhou Normal University, Hangzhou 311121, China; Department of Stomatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
| | - Liangjun Zhong
- Medical College of Hangzhou Normal University, Hangzhou 311121, China; Department of Stomatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China.
| |
Collapse
|
9
|
Zhang X, Liu Y, Kim YJ, Mac J, Zhuang R, Wang P. Co-delivery of carboplatin and paclitaxel via cross-linked multilamellar liposomes for ovarian cancer treatment. RSC Adv 2017; 7:19685-19693. [PMID: 28603607 PMCID: PMC5450007 DOI: 10.1039/c7ra01100h] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/30/2017] [Indexed: 02/06/2023] Open
Abstract
Cross-linked multilamellar liposomes offer an approach to achieve combinatorial delivery of hydrophobic paclitaxel and hydrophilic metallic carboplatin at a synergistic ratio to treat ovarian cancer.
Carboplatin (CPT) and paclitaxel (PTX) used in combination is one of the most effective treatments for ovarian cancer. However, the traditional combination methods used to co-administrate CPT and PTX showed limited clinical efficacy due to their distinct pharmacokinetics. Although much effort has been devoted to developing nanoparticles capable of encapsulating drugs with different lipophilicites, co-delivery of carboplatin with paclitaxel by a single nanoparticle has rarely been reported. Here, we encapsulated and delivered this drug combination to ovarian cancer cells at a controlled ratio by a previously reported crosslinked multilamellar liposome vesicle (cMLV). A 1 : 1 CPT/PTX molar ratio for cMLVs (CPT/PTX) combination treatment was found to induce the strongest anti-tumor synergism and to target ALDH+ cancer stem cells (CSC) in vitro. Moreover, we demonstrated that this co-encapsulation strategy reduced systemic cytotoxicity and resulted in a stronger anti-tumor effect when compared to free drug combinations and individual drug-loaded cMLVs in an OVCAR8 ovarian cancer xenograft mouse model. Thus, this study suggests a potentially promising combination therapy for ovarian cancer in clinical practice.
Collapse
Affiliation(s)
- Xiaoyang Zhang
- Mork Family Department of Chemical Engineering and Materials Science , University of Southern California , 3710 McClintock Ave. , RTH509 , Los Angeles , CA 90089 , USA . ; ; Tel: +1-213-740-0780
| | - Yarong Liu
- Mork Family Department of Chemical Engineering and Materials Science , University of Southern California , 3710 McClintock Ave. , RTH509 , Los Angeles , CA 90089 , USA . ; ; Tel: +1-213-740-0780
| | - Yu Jeong Kim
- Department of Pharmacology and Pharmaceutical Sciences , University of Southern California , Los Angeles , CA 90089 , USA
| | - John Mac
- Mork Family Department of Chemical Engineering and Materials Science , University of Southern California , 3710 McClintock Ave. , RTH509 , Los Angeles , CA 90089 , USA . ; ; Tel: +1-213-740-0780
| | - Rachel Zhuang
- Department of Biomedical Engineering , University of Southern California , Los Angeles , CA 90089 , USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science , University of Southern California , 3710 McClintock Ave. , RTH509 , Los Angeles , CA 90089 , USA . ; ; Tel: +1-213-740-0780.,Department of Pharmacology and Pharmaceutical Sciences , University of Southern California , Los Angeles , CA 90089 , USA.,Department of Biomedical Engineering , University of Southern California , Los Angeles , CA 90089 , USA
| |
Collapse
|
10
|
Huang Y, Yang X, Xu T, Kong Q, Zhang Y, Shen Y, Wei Y, Wang G, Chang KJ. Overcoming resistance to TRAIL-induced apoptosis in solid tumor cells by simultaneously targeting death receptors, c-FLIP and IAPs. Int J Oncol 2016; 49:153-63. [PMID: 27210546 PMCID: PMC4902065 DOI: 10.3892/ijo.2016.3525] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 04/15/2016] [Indexed: 12/12/2022] Open
Abstract
The discovery of the TRAIL protein and its death receptors DR4/5 changed the horizon of cancer research because TRAIL specifically kills cancer cells. However, the validity of TRAIL-based cancer therapies has yet to be established, as most cancer cells are TRAIL-resistant. In this report, we demonstrate that TRAIL-resistance of many cancer cell lines can be overcome after siRNA- or rocaglamide-mediated downregulation of c-FLIP expression and simultaneous inhibition of IAPs activity using AT406, a pan-antagonist of IAPs. Combined triple actions of the TRAIL, the IAPs inhibitor, AT406, and the c-FLIP expression inhibitor, rocaglamide (ART), markedly improve TRAIL-induced apoptotic effects in most solid cancer cell lines through the activation of an extrinsic apoptosis pathway. Furthermore, this ART combination does not harm normal cells. Among the 18 TRAIL-resistant cancer cell lines used, 15 cell lines become sensitive or highly sensitive to ART, and two out of three glioma cell lines exhibit high resistance to ART treatment due to very low levels of procaspase-8. This study provides a rationale for the development of TRAIL-induced apoptosis-based cancer therapies.
Collapse
Affiliation(s)
- Ying Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Xiang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Tianrui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Qinghong Kong
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Yaping Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Yuehai Shen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Yunlin Wei
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Guanlin Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Kwen-Jen Chang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
11
|
Liao W, Zhang R, Dong C, Yu Z, Ren J. Novel walnut peptide-selenium hybrids with enhanced anticancer synergism: facile synthesis and mechanistic investigation of anticancer activity. Int J Nanomedicine 2016; 11:1305-1321. [PMID: 27143875 PMCID: PMC4841427 DOI: 10.2147/ijn.s92257] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This contribution reports a facile synthesis of degreased walnut peptides (WP1)-functionalized selenium nanoparticles (SeNPs) hybrids with enhanced anticancer activity and a detailed mechanistic evaluation of its superior anticancer activity. Structural and chemical characterizations proved that SeNPs are effectively capped with WP1 via physical absorption, resulting in a stable hybrid structure with an average diameter of 89.22 nm. A panel of selected human cancer cell lines demonstrated high susceptibility toward WP1-SeNPs and displayed significantly reduced proliferative behavior. The as-synthesized WP1-SeNPs exhibited excellent selectivity between cancer cells and normal cells. The targeted induction of apoptosis in human breast adenocarcinoma cells (MCF-7) was confirmed by the accumulation of arrested S-phase cells, nuclear condensation, and DNA breakage. Careful investigations revealed that an extrinsic apoptotic pathway can be attributed to the cell apoptosis and the same was confirmed by activation of the Fas-associated with death domain protein and caspases 3, 8, and 9. In addition, it was also understood that intrinsic apoptotic pathways including reactive oxygen species generation, as well as the reduction in mitochondrial membrane potential, are also involved in the WP1-SeNP-induced apoptosis. This suggested the involvement of multiple apoptosis pathways in the anticancer activity. Our results indicated that WP1-SeNP hybrids with Se core encapsulated in a WP1 shell could be a highly effective method to achieve anticancer synergism. Moreover, the great potential exhibited by WP1-SeNPs could make them an ideal candidate as a chemotherapeutic agent for human cancers, especially for breast cancer.
Collapse
Affiliation(s)
- Wenzhen Liao
- College of Light Industry and Food Sciences, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Rong Zhang
- College of Light Industry and Food Sciences, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Chenbo Dong
- Civil and Environmental Engineering, Rice University, Houston, TX, USA
| | - Zhiqiang Yu
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Jiaoyan Ren
- College of Light Industry and Food Sciences, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
12
|
Capillary electrophoresis of RNA in hydroxyethylcellulose polymer with various molecular weights. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1011:114-20. [DOI: 10.1016/j.jchromb.2015.12.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/22/2015] [Accepted: 12/28/2015] [Indexed: 11/17/2022]
|
13
|
Sun W, Fang M, Chen Y, Yang Z, Xiao Y, Wan M, Wang H, Yu Y, Wang L. Delivery System of CpG Oligodeoxynucleotides through Eliciting an Effective T cell Immune Response against Melanoma in Mice. J Cancer 2016; 7:241-50. [PMID: 26918036 PMCID: PMC4747877 DOI: 10.7150/jca.12899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 11/13/2015] [Indexed: 01/22/2023] Open
Abstract
Purpose: In order to improve the immunogenicity of whole tumor cell lysate for tumor vaccine, we have designed a series of CpG ODNs to study their transport and to evaluate their anti-tumor activity in B16 melanoma mouse models. Methods: In this study, we investigated whether C-class CpG ODN (CpG ODN-685) could facilitate tumor cell lysate to induce vigorous anti-tumor activity against tumors in mice both prophylactically and therapeutically. Results: It was found that the combination of tumor cell lysate and CpG ODN-685 could inhibit the growth of B16 melanoma and prolong the survival of tumor-bearing mice. Moreover CpG ODN-685 with the addition of tumor cell lysate can also cause the generation of tumor specific immune memory by inducing specific cytotoxic T lymphocytes and helper T lymphocytes in mice. Conclusion: The results suggest that CpG ODN-685 could be developed as an efficient adjuvant for tumor vaccines against melanoma.
Collapse
Affiliation(s)
- Wei Sun
- 1. Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Mingli Fang
- 1. Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yajing Chen
- 1. Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Zhaogang Yang
- 3. NSF Nanoscale Science and Engineering Center (NSEC), The Ohio State University, Columbus, OH 43212, USA
| | - Yue Xiao
- 1. Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Min Wan
- 1. Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Hua Wang
- 1. Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yongli Yu
- 2. Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Liying Wang
- 1. Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
14
|
Yu B, Sun XN, Shi XJ, Qi PP, Zheng YC, Yu DQ, Liu HM. Efficient synthesis of novel antiproliferative steroidal spirooxindoles via the [3+2] cycloaddition reactions of azomethine ylides. Steroids 2015; 102:92-100. [PMID: 26256638 DOI: 10.1016/j.steroids.2015.08.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 08/04/2015] [Indexed: 12/31/2022]
Abstract
A series of novel steroidal spirooxindoles 3a-h were synthesized from pregnenolone in a high regioselective manner using the 1,3-dipolar cycloaddition as the key step. This protocol resulted in the formation of two C-C bonds, one C-N bond and the creation of one pyrrolidine ring and three contiguous stereocenters in a single operation. Biological evaluation showed that these synthesized steroidal spirooxindoles exhibited moderate to good antiproliferative activity against the tested cell lines and some of them were more potent than 5-FU. Among them, compounds 3e and 3f displayed the best antiproliferative activity against MCF-7 cells with the IC50 values of 4.0 and 3.9μM, respectively. Flow cytometry analysis demonstrated that compound 3d caused the cellular apoptosis and cell cycle arrest at G2/M phase in a concentration-dependent manner. Docking results indicated that compound 3d fitted well into the MDM2 active site 1RV1 by interacting with Lys94 and Thr101 residues.
Collapse
Affiliation(s)
- Bin Yu
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Xiao-Nan Sun
- The Affiliated Hospital of Huanghe Sanmenxia of Henan University of Science and Technology, Sanmenxia 472000, PR China
| | - Xiao-Jing Shi
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ping-Ping Qi
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yi-Chao Zheng
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China
| | - De-Quan Yu
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Hong-Min Liu
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
15
|
Li Y, Liu D, Zhou Y, Li Y, Xie J, Lee RJ, Cai Y, Teng L. Silencing of Survivin Expression Leads to Reduced Proliferation and Cell Cycle Arrest in Cancer Cells. J Cancer 2015; 6:1187-94. [PMID: 26516368 PMCID: PMC4615356 DOI: 10.7150/jca.12437] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/27/2015] [Indexed: 01/20/2023] Open
Abstract
Survivin is an anti-apoptotic gene that is overexpressed in most human tumors. RNA interference using short interfering RNA (siRNA) can be used to specifically inhibit survivin expression. Tumor cells were treated with a newly designed survivin siRNA, which was modified with 2′-OMe. Cellular survivin mRNA and protein levels were determined by real-time qRT-PCR and Western blot, respectively. Cell cycle and apoptosis were determined by flow cytometry. Cell proliferation was measured by MTT assay. Our data showed that the novel survivin-targeted siRNA could efficiently knockdown the expression of survivin and inhibit cell proliferation. Survivin mRNA was reduced by 95% after 48h treatment with 20nM siRNA. In addition, the siRNA could markedly arrest the cell cycle at the G2/M checkpoint and induce cellular apoptosis in a dose-dependent manner. The percentage of apoptotic cells reached 50% when treated with 40nM siRNA. In conclusion, we have identified a novel chemically modified siRNA against survivin that is highly efficient and delineated its mechanism of action, thus demonstrating a potential therapeutic role for this molecule in cancer. Further evaluation of this siRNA for therapeutic activity is warranted.
Collapse
Affiliation(s)
- Yuhuan Li
- 1. Institute of Life Sciences, Jilin University, Changchun, Jilin, P. R. China
| | - Da Liu
- 1. Institute of Life Sciences, Jilin University, Changchun, Jilin, P. R. China
| | - Yulin Zhou
- 1. Institute of Life Sciences, Jilin University, Changchun, Jilin, P. R. China
| | - Yujing Li
- 1. Institute of Life Sciences, Jilin University, Changchun, Jilin, P. R. China
| | - Jing Xie
- 1. Institute of Life Sciences, Jilin University, Changchun, Jilin, P. R. China
| | - Robert J Lee
- 1. Institute of Life Sciences, Jilin University, Changchun, Jilin, P. R. China ; 2. Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH, U.S.A
| | - Yong Cai
- 1. Institute of Life Sciences, Jilin University, Changchun, Jilin, P. R. China
| | - Lesheng Teng
- 1. Institute of Life Sciences, Jilin University, Changchun, Jilin, P. R. China
| |
Collapse
|
16
|
Zhu J, Wang J, Wang X, Zhu J, Yang Y, Tian J, Cui W, Ge C, Li Y, Pan Y, Gu H. Facile synthesis of magnetic core-shell nanocomposites for MRI and CT bimodal imaging. J Mater Chem B 2015; 3:6905-6910. [PMID: 32262539 DOI: 10.1039/c5tb00775e] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
With the development of nanotechnology, nanocomposites have been used as bimodal contrast agents for magnetic resonance (MR) and computed tomography (CT) imaging. We have developed a facile method for the synthesis of iron oxide@bismuth sulfide magnetic core-shell nanocomposites. These bifunctional nanocomposites can be made water-soluble via PEG coating and present strong MRI/CT contrast enhancement. Evaluation of cytotoxicity by MTT assay shows that the nanocomposites have low cytotoxicity. The results illustrate that the nanocomposites have great potential as bimodal imaging agents for MR/CT.
Collapse
Affiliation(s)
- Jing Zhu
- Key Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, 215123, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Shi YQ, Yan CC, Zhang X, Yan M, Liu LR, Geng HZ, Lv L, Li BX. Mechanisms underlying probucol-induced hERG-channel deficiency. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3695-704. [PMID: 26229434 PMCID: PMC4516208 DOI: 10.2147/dddt.s86724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The hERG gene encodes the pore-forming α-subunit of the rapidly activating delayed rectifier potassium channel (I Kr), which is important for cardiac repolarization. Reduction of I hERG due to genetic mutations or drug interferences causes long QT syndrome, leading to life-threatening cardiac arrhythmias (torsades de pointes) or sudden death. Probucol is a cholesterol-lowering drug that could reduce hERG current by decreasing plasma membrane hERG protein expression and eventually cause long QT syndrome. Here, we investigated the mechanisms of probucol effects on I hERG and hERG-channel expression. Our data demonstrated that probucol reduces SGK1 expression, known as SGK isoform, in a concentration-dependent manner, resulting in downregulation of phosphorylated E3 ubiquitin ligase Nedd4-2 expression, but not the total level of Nedd4-2. As a result, the hERG protein reduces, due to the enhanced ubiquitination level. On the contrary, carbachol could enhance the phosphorylation level of Nedd4-2 as an alternative to SGK1, and thus rescue the ubiquitin-mediated degradation of hERG channels caused by probucol. These discoveries provide a novel mechanism of probucol-induced hERG-channel deficiency, and imply that carbachol or its analog may serve as potential therapeutic compounds for the handling of probucol cardiotoxicity.
Collapse
Affiliation(s)
- Yuan-Qi Shi
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Cai-Chuan Yan
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xiao Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Meng Yan
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Li-Rong Liu
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Huai-Ze Geng
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Lin Lv
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Bao-Xin Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China ; State-Province Key Laboratory of Biopharmaceutical Engineering, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
18
|
Kim YJ, Liu Y, Li S, Rohrs J, Zhang R, Zhang X, Wang P. Co-Eradication of Breast Cancer Cells and Cancer Stem Cells by Cross-Linked Multilamellar Liposomes Enhances Tumor Treatment. Mol Pharm 2015; 12:2811-22. [PMID: 26098197 DOI: 10.1021/mp500754r] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The therapeutic limitations of conventional chemotherapeutic drugs have emerged as a challenge for breast cancer therapy; these shortcomings are likely due, at least in part, to the presence of the cancer stem cells (CSCs). Salinomycin, a polyether antibiotic isolated from Streptomyces albus, has been shown to selectively inhibit cancer stem cells; however, its clinical application has been hindered by the drug's hydrophobility, which limits the available administration routes. In this paper, a novel drug delivery system, cross-linked multilamellar liposomal vesicles (cMLVs), was optimized to allow for the codelivery of salinomycin (Sal) and doxorubicin (Dox), targeting both CSCs and breast cancer cells. The results show that the cMLV particles encapsulating different drugs have similar sizes with high encapsulation efficiencies (>80%) for both Dox and Sal. Dox and Sal were released from the particles in a sustained manner, indicating the stability of the cMLVs. Moreover, the inhibition of cMLV(Dox+Sal) against breast cancer cells was stronger than either single-drug treatment. The efficient targeting of cMLV(Dox+Sal) to CSCs was validated through in vitro experiments using breast cancer stem cell markers. In accordance with the in vitro combination treatment, in vivo breast tumor suppression by cMLV(Dox+Sal) was 2-fold more effective than single-drug cMLV treatment or treatment with the combination of cMLV(Dox) and cMLV(Sal). Thus, this study demonstrates that cMLVs represent a novel drug delivery system that can serve as a potential platform for combination therapy, allowing codelivery of an anticancer agent and a CSC inhibitor for the elimination of both breast cancer cells and cancer stem cells.
Collapse
Affiliation(s)
- Yu Jeong Kim
- †Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Yarong Liu
- ‡Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| | - Si Li
- †Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States
| | - Jennifer Rohrs
- §Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Rachel Zhang
- §Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Xiaoyang Zhang
- ‡Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
| | - Pin Wang
- †Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, California 90089, United States.,‡Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States.,§Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
19
|
Zhang H, Patel N, Xiong J, Ding S. Targeting and noninvasive treatment of hepatocellular carcinoma in situ by ZnO nanorod-mediated concurrent chemoradiotherapy. RSC Adv 2015; 5:85720-85729. [DOI: 10.1039/c5ra16880e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
TfR Ab/Dox/ZnO nanocomposites, in which transferrin receptor antibody (TfR Ab) functionalized ZnO nanorods, loaded with doxorubicin (Dox), were prepared to mediate concurrent chemoradiotherapy for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Haijun Zhang
- Department of Oncology
- Zhongda Hospital
- Medical School
- Southeast University
- Nanjing
| | - Nishant Patel
- Department of Oncology
- Zhongda Hospital
- Medical School
- Southeast University
- Nanjing
| | - Jian Xiong
- Department of Oncology
- Zhongda Hospital
- Medical School
- Southeast University
- Nanjing
| | - Shuang Ding
- Department of Oncology
- Zhongda Hospital
- Medical School
- Southeast University
- Nanjing
| |
Collapse
|