1
|
Jin M, Cho HJ, Na YG, Yun TS, Song B, Lee SR, Je S, Oh HG, Park YG, Rho J, Yang MJ, Shin JS, Baek JS, Lee HK, Kim TW, Cho CW. Practical approach to development of GS-445124-loaded PLGA nanoparticles for the long-term treatment of feline infectious peritonitis caused by feline coronavirus infection. Int J Pharm 2025; 674:125468. [PMID: 40107469 DOI: 10.1016/j.ijpharm.2025.125468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/13/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Feline infectious peritonitis virus (FIPV) caused by feline coronavirus (FCoV) infection leads to a high mortality rate when untreated. GS-441524, an antiviral agent effective against FIPV, is orally administered twice daily or through daily subcutaneous injections for approximately 12 weeks. While the short treatment period recuses concerns about adherence, frequent administrations may cause handling-related stress in cats. Therefore, it is essential to develop a long-acting formulation that requires only a single administration. In this study, we used polylactide-co-glycolide (PLGA) as a carrier, which was used to effectively encapsulate GS-441524 with sustained-release functionality and GS-441524-loaded PLGA nanoparticles (GS-PLGA NP) were prepared. The particle size of GS-PLGA NP was 216 nm, the encapsulation efficiency was 78 %, and the 7-day release was 92 %. When GS-PLGA NP was injected at 22 mg/kg in cats, higher systemic exposure can be expected compared to injecting GS-441524 at 4 mg/kg for one week (relative bioavailability, 152 %). As well as GS-PLGA NP showed lower toxicity, improved cellular uptake, and enhanced antiviral efficacy against FCoV compared to the pure GS-441524.
Collapse
Affiliation(s)
- Minki Jin
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Hyun-Jin Cho
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea; College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Young-Guk Na
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Taek-Seon Yun
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Bomin Song
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Sang-Rae Lee
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Seonho Je
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Hong-Geun Oh
- R&D Division, Huvet Co., Ltd, Iksan-si 54541, Republic of Korea
| | - Yang-Gyu Park
- R&D Division, Huvet Co., Ltd, Iksan-si 54541, Republic of Korea
| | - Jinhyung Rho
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Mi-Jin Yang
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, Jeongeup, 56212, Republic of Korea
| | - Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jong-Suep Baek
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hong-Ki Lee
- College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-Gu, Cheongju, Chungbuk 28644, Republic of Korea.
| | - Tae-Won Kim
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| | - Cheong-Weon Cho
- College of Pharmacy and Institute of Drug Research and Development Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
2
|
Fan WL, Huang SY, Yang XJ, Bintang Ilhami F, Chen JK, Cheng CC. Hydrogen-bonded cytosine-endowed supramolecular polymeric nanogels: Highly efficient cancer cell targeting and enhanced therapeutic efficacy. J Colloid Interface Sci 2024; 665:329-344. [PMID: 38531278 DOI: 10.1016/j.jcis.2024.03.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/10/2024] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
We demonstrate that cytosine moieties within physically cross-linked supramolecular polymers not only manipulate drug delivery and release, but also confer specific targeting of cancer cells to effectively enhance the safety and efficacy of chemotherapy-and thus hold significant potential as a new perspective for development of drug delivery systems. Herein, we successfully developed physically cross-linked supramolecular polymers (PECH-PEG-Cy) comprised of hydrogen-bonding cytosine pendant groups, hydrophilic poly(ethylene glycol) side chains, and a hydrophobic poly(epichlorohydrin) main chain. The polymers spontaneously self-assemble into a reversibly hydrogen-bonded network structure induced by cytosine and directly form spherical nanogels in aqueous solution. Nanogels with a high hydrogen-bond network density (i.e., a higher content of cytosine moieties) exhibit outstanding long-term structural stability in cell culture substrates containing serum, whereas nanogels with a relatively low hydrogen-bond network density cannot preserve their structural integrity. The nanogels also exhibit numerous unique physicochemical characteristics in aqueous solution, such as a desirable spherical size, high biocompatibility with normal and cancer cells, excellent drug encapsulation capacity, and controlled pH-responsive drug release properties. More importantly, in vitro experiments conclusively indicate the drug-loaded PECH-PEG-Cy nanogels can selectively induce cancer cell-specific apoptosis and cell death via cytosine receptor-mediated endocytosis, without significantly harming normal cells. In contrast, control drug-loaded PECH-PEG nanogels, which lack cytosine moieties in their structure, can only induce cell death in cancer cells through non-specific pathways, which significantly inhibits the induction of apoptosis. This work clearly demonstrates that the cytosine moieties in PECH-PEG-Cy nanogels confer selective affinity for the surface of cancer cells, which enhances their targeted cellular uptake, cytotoxicity, and subsequent induction of programmed cell death in cancer cells.
Collapse
Affiliation(s)
- Wen-Lu Fan
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Shan-You Huang
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Xiu-Jing Yang
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Fasih Bintang Ilhami
- Department of Natural Science, Faculty of Mathematics and Natural Science, Universitas Negeri Surabaya, Surabaya 60231, Indonesia
| | - Jem-Kun Chen
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Chih-Chia Cheng
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan; Advanced Membrane Materials Research Center, National Taiwan University of Science and Technology, Taipei 10607, Taiwan.
| |
Collapse
|
3
|
Meng X, Zhu G, Yang YG, Sun T. Targeted delivery strategies: The interactions and applications of nanoparticles in liver diseases. Biomed Pharmacother 2024; 175:116702. [PMID: 38729052 DOI: 10.1016/j.biopha.2024.116702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
In recent years, nanoparticles have been broadly utilized in various drugs delivery formulations. Nanodelivery systems have shown promise in solving problems associated with the distribution of hydrophobic drugs and have promoted the accumulation of nanomedicines in the circulation or in organs. However, the injection dose of nanoparticles (NPs) is much greater than that needed by diseased tissues or organs. In other words, most of the NPs are localized off-target and do not reach the desired tissue or organs. With the rapid development of biodegradable and biosafety nanomaterials, the nanovectors represent assurance of safety. However, the off-target effects also induce concerns about the application of NPs, especially in the delivery of gene editing tools. Therefore, a complete understanding of the biological responses to NPs in the body will clearly guide the design of targeted delivery of NPs. The different properties of various nanodelivery systems may induce diverse interactions between carriers and organs. In this review, we describe the relationship between the liver, the most influenced organ of systemic administration of NPs, and targeted delivery nanoplatforms. Various transport vehicles have adopted multiple delivery strategies for the targeted delivery to the cells in the homeostasis liver and in diseased liver. Additionally, nanodelivery systems provide a novel strategy for treating incurable diseases. The appearance of a targeted delivery has profoundly improved the application of NPs to liver diseases.
Collapse
Affiliation(s)
- Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Ge Zhu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
4
|
Lu J, Dai Y, He Y, Zhang T, Zhang J, Chen X, Jiang C, Lu H. Organ/Cell-Selective Intracellular Delivery of Biologics via N-Acetylated Galactosamine-Functionalized Polydisulfide Conjugates. J Am Chem Soc 2024; 146:3974-3983. [PMID: 38299512 DOI: 10.1021/jacs.3c11914] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Biologics, including proteins and antisense oligonucleotides (ASOs), face significant challenges when it comes to achieving intracellular delivery within specific organs or cells through systemic administrations. In this study, we present a novel approach for delivering proteins and ASOs to liver cells, both in vitro and in vivo, using conjugates that tether N-acetylated galactosamine (GalNAc)-functionalized, cell-penetrating polydisulfides (PDSs). The method involves the thiol-bearing cargo-mediated ring-opening polymerization of GalNAc-functionalized lipoamide monomers through the so-called aggregation-induced polymerization, leading to the formation of site-specific protein/ASO-PDS conjugates with narrow dispersity. The hepatocyte-selective intracellular delivery of the conjugates arises from a combination of factors, including first GalNAc binding with ASGPR receptors on liver cells, leading to cell immobilization, and the subsequent thiol-disulfide exchange occurring on the cell surface, promoting internalization. Our findings emphasize the critical role of the close proximity of the PDS backbone to the cell surface, as it governs the success of thiol-disulfide exchange and, consequently, cell penetration. These conjugates hold tremendous potential in overcoming the various biological barriers encountered during systemic and cell-specific delivery of biomacromolecular cargos, opening up new avenues for the diagnosis and treatment of a range of liver-targeting diseases.
Collapse
Affiliation(s)
- Jianhua Lu
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, People's Republic of China
| | - Yuanhao Dai
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, People's Republic of China
| | - Yahui He
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, People's Republic of China
| | - Ting Zhang
- Department of Microbiology & Infectious Disease Center, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, People's Republic of China
| | - Jing Zhang
- Department of Microbiology & Infectious Disease Center, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, People's Republic of China
| | - Xiangmei Chen
- Department of Microbiology & Infectious Disease Center, Peking University Health Science Center, 38 Xueyuan Road, Beijing 100191, People's Republic of China
| | - Changtao Jiang
- Department of Immunology, School of Basic Medical Sciences, State Key Laboratory of Female Fertility Promotion, Peking University, Beijing 100191, China
| | - Hua Lu
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, People's Republic of China
| |
Collapse
|
5
|
Huang R, Wang F, Fu H, Qi X, Xing G, Ren J, Cheng L, Meng F, Zhong Z. Bioresponsive Chimaeric Polymersomes Mediate Sustained and Liver-Specific siRNA Transfection In Vivo. Biomacromolecules 2023; 24:5353-5363. [PMID: 37871289 DOI: 10.1021/acs.biomac.3c00813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The silencing of disease-causing genes with small interfering RNA (siRNA) offers a particularly effective therapeutic strategy for different disorders; however, its clinical efficacy relies on the development of nontoxic and tissue-specific delivery vehicles. Herein, we report that bioresponsive chimaeric polymersomes (BCP) with short poly(ethylenimine) as inner shell mediate highly efficacious, sustained, and liver-specific siRNA transfection in vivo. BCP exhibited remarkable encapsulation efficiencies of siRNA (95-100%) at siRNA-feeding contents of 15-25 wt %, to afford stable, small-sized (55-64 nm), and neutral-charged BCP-siRNA. siApoB-Loaded BCP (BCP-siApoB) outperformed lipofectamine counterparts and silenced 93% of ApoB mRNA in HepG2 cells at 50 nM siApoB without inducing cytotoxicity. Intriguingly, the in vivo studies using wild-type C57BL/6 mice revealed that BCP-siApoB preferentially accumulated in the liver, and a single dose of 4.5 mg/kg achieved over 90% downregulation of ApoB mRNA for at least 10 days. The systemic administration of BCP-siApoB at 4.5 mg/kg every 2 weeks or 1.5 mg/kg weekly in diet-induced obese mice could also achieve up to 80% silencing of ApoB mRNA. The liver specificity and silencing efficacy of BCP-siApoB could further be improved by decorating it with the trivalent N-acetylgalactosamine (TriGalNAc) ligand. These bioresponsive and liver-specific chimaeric polymersomes provide an enabling technology for siRNA therapy of various liver-related diseases.
Collapse
Affiliation(s)
- Ri Huang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Feifei Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - He Fu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Xinming Qi
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Guozhen Xing
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P. R. China
| | - Liang Cheng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
6
|
Han S, Chi Y, Yang Z, Ma J, Wang L. Tumor Microenvironment Regulation and Cancer Targeting Therapy Based on Nanoparticles. J Funct Biomater 2023; 14:136. [PMID: 36976060 PMCID: PMC10053410 DOI: 10.3390/jfb14030136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Although we have made remarkable achievements in cancer awareness and medical technology, there are still tremendous increases in cancer incidence and mortality. However, most anti-tumor strategies, including immunotherapy, show low efficiency in clinical application. More and more evidence suggest that this low efficacy may be closely related to the immunosuppression of the tumor microenvironment (TME). The TME plays a significant role in tumorigenesis, development, and metastasis. Therefore, it is necessary to regulate the TME during antitumor therapy. Several strategies are developing to regulate the TME as inhibiting tumor angiogenesis, reversing tumor associated macrophage (TAM) phenotype, removing T cell immunosuppression, and so on. Among them, nanotechnology shows great potential for delivering regulators into TME, which further enhance the antitumor therapy efficacy. Properly designed nanomaterials can carry regulators and/or therapeutic agents to eligible locations or cells to trigger specific immune response and further kill tumor cells. Specifically, the designed nanoparticles could not only directly reverse the primary TME immunosuppression, but also induce effective systemic immune response, which would prevent niche formation before metastasis and inhibit tumor recurrence. In this review, we summarized the development of nanoparticles (NPs) for anti-cancer therapy, TME regulation, and tumor metastasis inhibition. We also discussed the prospect and potential of nanocarriers for cancer therapy.
Collapse
Affiliation(s)
- Shulan Han
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Yongjie Chi
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhu Yang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juan Ma
- Department of Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Lianyan Wang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
7
|
Athanasopoulou F, Manolakakis M, Vernia S, Kamaly N. Nanodrug delivery systems for metabolic chronic liver diseases: advances and perspectives. Nanomedicine (Lond) 2023; 18:67-84. [PMID: 36896958 DOI: 10.2217/nnm-2022-0261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/03/2023] [Indexed: 03/11/2023] Open
Abstract
Nanomedicines are revolutionizing healthcare as recently demonstrated by the Pfizer/BioNTech and Moderna COVID-2019 vaccines, with billions of doses administered worldwide in a safe manner. Nonalcoholic fatty liver disease is the most common noncommunicable chronic liver disease, posing a major growing challenge to global public health. However, due to unmet diagnostic and therapeutic needs, there is great interest in the development of novel translational approaches. Nanoparticle-based approaches offer novel opportunities for efficient and specific drug delivery to liver cells, as a step toward precision medicines. In this review, the authors highlight recent advances in nanomedicines for the generation of novel diagnostic and therapeutic tools for nonalcoholic fatty liver disease and related liver diseases.
Collapse
Affiliation(s)
- Foteini Athanasopoulou
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, W12 0BZ, UK
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Michail Manolakakis
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, W12 0BZ, UK
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Santiago Vernia
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Nazila Kamaly
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, W12 0BZ, UK
| |
Collapse
|
8
|
Zhou Y, Li S, Tan W, Wei X, Chang Y, Yi Q, Pu C, Wang J. Design, Synthesis, and Preparation of Ultrasound-Responsive Curcumin-Loaded Chitosan Nanocarriers. RUSS J GEN CHEM+ 2023. [DOI: 10.1134/s1070363223010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
9
|
Kakavand H, Aghakouchakzadeh M, Shahi A, Virani SS, Dixon DL, Van Tassell BW, Talasaz AH. A stepwise approach to prescribing novel lipid-lowering medications. J Clin Lipidol 2022; 16:822-832. [DOI: 10.1016/j.jacl.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/19/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
|
10
|
Square wave voltammetric approach to leptin immunosensing and optimization of driving parameters with chemometrics. Biosens Bioelectron 2022; 216:114592. [PMID: 35969964 DOI: 10.1016/j.bios.2022.114592] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 12/28/2022]
Abstract
Square wave voltammetry serves as an effective analytical means to evaluate antigen-antibody coupling at the solid-liquid interface. Herein, we describe 3-aminopropyltrimethoxysilane (APTMS) induced irreversible immobilization of anti-leptin to micellar gold nanoparticles (AuNPs). Antibodies (Abs) were orthogonally loaded on micellized AuNP assemblies via amino residual groups. The ratio of bound Ab molecules was determined by the Bradford assay. The AuNP/Ab layer modified electrodes with variable antibody surface coverage (∼400 ± 55-200 ± 30 Ab/NP) were analyzed in terms of change in backward, net current (Ip) components. The rate of antigen coupling was found to be consistent with the variation in antibody density as well as the binding affinity. The lowest detection limit was observed at the femtomolar level (0.25 fM/mL) over a wide range of antigen concentration (6.2 ng/mL to 0.12 fg/mL). The variables affecting the epitope-paratope interaction were further optimized using a chemometric approach and a response surface methodology (RSM).
Collapse
|
11
|
Miao J, Gao P, Li Q, He K, Zhang L, Wang J, Huang L. Advances in Nanoparticle Drug Delivery Systems for Anti-Hepatitis B Virus Therapy: A Narrative Review. Int J Mol Sci 2021; 22:ijms222011227. [PMID: 34681886 PMCID: PMC8538950 DOI: 10.3390/ijms222011227] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/26/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic hepatitis B (CHB) is an infectious viral disease that is prevalent worldwide. Traditional nucleoside analogues, as well as the novel drug targets against hepatitis B virus (HBV), are associated with certain critical factors that influence the curative effect, such as biological stability and safety, effective drug delivery, and controlled release. Nanoparticle drug delivery systems have significant advantages and have provided a basis for the development of anti-HBV strategies. In this review, we aim to review the advances in nanoparticle drug delivery systems for anti-hepatitis B virus therapy by summarizing the relevant literature. First, we focus on the characteristics of nanoparticle drug delivery systems for anti-HBV therapy. Second, we discuss the nanoparticle delivery systems for anti-HBV nucleoside drugs, gene-based drugs, and vaccines. Lastly, we provide an overview of the prospects for nanoparticle-based anti-HBV agents.
Collapse
Affiliation(s)
- Jing Miao
- Department of Pharmacy, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; (J.M.); (P.G.); (K.H.); (L.Z.)
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Hangzhou 310003, China
| | - Peng Gao
- Department of Pharmacy, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; (J.M.); (P.G.); (K.H.); (L.Z.)
| | - Qian Li
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China;
| | - Kaifeng He
- Department of Pharmacy, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; (J.M.); (P.G.); (K.H.); (L.Z.)
| | - Liwen Zhang
- Department of Pharmacy, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; (J.M.); (P.G.); (K.H.); (L.Z.)
| | - Junyan Wang
- Department of Pharmacy, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; (J.M.); (P.G.); (K.H.); (L.Z.)
- Correspondence: (J.W.); (L.H.)
| | - Lingfei Huang
- Department of Pharmacy, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China; (J.M.); (P.G.); (K.H.); (L.Z.)
- Correspondence: (J.W.); (L.H.)
| |
Collapse
|
12
|
Li T, Zhu L, Zhu L, Wang P, Xu W, Huang J. Recent Developments in Delivery of MicroRNAs Utilizing Nanosystems for Metabolic Syndrome Therapy. Int J Mol Sci 2021; 22:ijms22157855. [PMID: 34360621 PMCID: PMC8346175 DOI: 10.3390/ijms22157855] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic syndrome (MetS) is a set of complex, chronic inflammatory conditions that are characterized by central obesity and associated with an increased risk of cardiovascular diseases. In recent years, microRNAs (miRNAs) have become an important type of endocrine factors, which play crucial roles in maintaining energy balance and metabolic homeostasis. However, its unfavorable properties such as easy degradation in blood and off-target effect are still a barrier for clinical application. Nanosystem based delivery possess strong protection, high bioavailability and control release rate, which is beneficial for success of gene therapy. This review first describes the current progress and advances on miRNAs associated with MetS, then provides a summary of the therapeutic potential and targets of miRNAs in metabolic organs. Next, it discusses recent advances in the functionalized development of classic delivery systems (exosomes, liposomes and polymers), including their structures, properties, functions and applications. Furthermore, this work briefly discusses the intelligent strategies used in emerging novel delivery systems (selenium nanoparticles, DNA origami, microneedles and magnetosomes). Finally, challenges and future directions in this field are discussed provide a comprehensive overview of the future development of targeted miRNAs delivery for MetS treatment. With these contributions, it is expected to address and accelerate the development of effective NA delivery systems for the treatment of MetS.
Collapse
Affiliation(s)
- Tong Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (T.L.); (L.Z.); (L.Z.); (P.W.); (W.X.)
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Liye Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (T.L.); (L.Z.); (L.Z.); (P.W.); (W.X.)
| | - Longjiao Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (T.L.); (L.Z.); (L.Z.); (P.W.); (W.X.)
| | - Pengjie Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (T.L.); (L.Z.); (L.Z.); (P.W.); (W.X.)
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (T.L.); (L.Z.); (L.Z.); (P.W.); (W.X.)
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Jiaqiang Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (T.L.); (L.Z.); (L.Z.); (P.W.); (W.X.)
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
- Correspondence:
| |
Collapse
|
13
|
Tieu T, Wei Y, Cifuentes‐Rius A, Voelcker NH. Overcoming Barriers: Clinical Translation of siRNA Nanomedicines. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100108] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Terence Tieu
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
| | - Yingkai Wei
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Anna Cifuentes‐Rius
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Nicolas H. Voelcker
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
- Melbourne Centre for Nanofabrication 151 Wellington Road Victorian Node of the Australian National Fabrication Facility Clayton VIC 3168 Australia
| |
Collapse
|
14
|
Saadat M, Mostafaei F, Mahdinloo S, Abdi M, Zahednezhad F, Zakeri-Milani P, Valizadeh H. Drug delivery of pH-Sensitive nanoparticles into the liver cancer cells. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102557] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
15
|
Fleischmann D, Goepferich A. General sites of nanoparticle biodistribution as a novel opportunity for nanomedicine. Eur J Pharm Biopharm 2021; 166:44-60. [PMID: 34087354 DOI: 10.1016/j.ejpb.2021.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/22/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
The development of nanomedical devices has led to a considerable number of clinically applied nanotherapeutics. Yet, the overall poor translation of nanoparticular concepts into marketable systems has not met the initial expectations and led to increasing criticism in recent years. Most novel nano approaches thereby use highly refined formulations including a plethora of active targeting sequences, but ultimately fail to reach their target due to a generally high off-target deposition in organs such as the liver or kidney. In this context, we argue that initial nanoparticle (NP) development should not entirely become set on conventional formulation aspects. In contrast, we propose a change of focus towards a prior analysis of general sites of NP in vivo deposition and an assessment of how accumulation in these organs or tissues can be harnessed to develop therapies for site-related pathologies. We therefore give a comprehensive overview of existing nanotherapeutic targeting strategies for specific cell types within three of the usual suspects, i.e. the liver, kidney and the vascular system. We discuss the physiological surroundings and relevant pathologies of described tissues as well as the implications for NP-mediated drug delivery. Additionally, successful cell-selective NP concepts using active targeting strategies are assessed. By bringing together both (patho)physiological aspects and concepts for cell-selective NP formulations, we hope to show a novel opportunity for the development of more promising nanotherapeutic devices.
Collapse
Affiliation(s)
- Daniel Fleischmann
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany.
| |
Collapse
|
16
|
Wang Y, Ding R, Zhang Z, Zhong C, Wang J, Wang M. Curcumin-loaded liposomes with the hepatic and lysosomal dual-targeted effects for therapy of hepatocellular carcinoma. Int J Pharm 2021; 602:120628. [PMID: 33892061 DOI: 10.1016/j.ijpharm.2021.120628] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/20/2021] [Accepted: 04/17/2021] [Indexed: 12/13/2022]
Abstract
Curcumin can induce cancer cell apoptosis through lysosomal permeabilization pathway. However, the poor selectivity of curcumin restricts its use in the therapy of hepatocellular carcinoma. Because galactose group can recognize ASGPR overexpressed on hepatoma cells and morpholine group can target to the lysosome, they are integrated into a dual-targeted lipid material with low toxicity. The corresponding galactose-morpholine modified liposomes loaded with curcumin (Gal-Mor-LPs) were prepared and evaluated in comparison with conventional liposomes (LPs) and galactose modified liposomes (Gal-LPs). The in vitro and in vivo hepatic targeting capacity of liposomes followed a trend of LPs < Gal-LPs < Gal-Mor-LPs. The endocytosis of Gal-Mor-LPs was competitively inhibited by galactose, which confirmed the galactose modified liposomes entered hepatoma cells via ASGPR-mediated pathway. Gal-Mor-LPs displayed more excellent lysosomal targeting efficacy than LPs and Gal-LPs due to the attraction of acidic lysosome on basic morpholine group of Gal-Mor-LPs. The in vivo tumor inhibition effects of formulations also followed a trend of free curcumin < LPs < Gal-LPs < Gal-Mor-LPs, confirming that hepatic and lysosomal dual-targeting vehicle can improve the antitumor efficacy of curcumin. Moreover, the curcumin-loaded liposomes modified with galactose and morpholine moieties show good biocompatibility in vivo.
Collapse
Affiliation(s)
- Yan Wang
- College of Life Science and Technology, Guangxi University, Nanning 530004, PR China
| | - Ruihua Ding
- College of Life Science and Technology, Guangxi University, Nanning 530004, PR China; Medical College, Guangxi University, Nanning 530004, PR China
| | - Zan Zhang
- Guangxi Institute for Food and Drug Control, Nanning 530021, PR China
| | - Cheng Zhong
- School of Computer, Electronics and Information, Guangxi University, Nanning 530004, PR China
| | - Jianyi Wang
- Medical College, Guangxi University, Nanning 530004, PR China.
| | - Mian Wang
- College of Life Science and Technology, Guangxi University, Nanning 530004, PR China.
| |
Collapse
|
17
|
Miao J, Yang X, Shang X, Gao Z, Li Q, Hong Y, Wu J, Meng T, Yuan H, Hu F. Hepatocyte-targeting and microenvironmentally responsive glycolipid-like polymer micelles for gene therapy of hepatitis B. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:127-139. [PMID: 33738144 PMCID: PMC7943969 DOI: 10.1016/j.omtn.2021.02.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022]
Abstract
Hepatitis B (HB) is a viral infectious disease that seriously endangers human health, and since there are no radical drugs to counter this, effective and safe therapies urgently need to be developed. HB virus (HBV) mainly infects hepatocytes (HCs), while the drugs are easily phagocytosed by Kupffer cells (KCs). In this study, the glutathione concentration difference between HCs and KCs was examined and utilized in an ideal drug-release strategy. Here, galactosylated chitosan-oligosaccharide-SS-octadecylamine (Gal-CSSO) was prepared to accurately deliver 10-23 DNAzyme DrzBC (blocking HBeAg expression) or DrzBS (blocking HBsAg expression) in targeted HB therapy. In vitro Gal-CSSO systems exhibited low cytotoxicity, endosomal escape, and glutathione responsiveness. The HBeAg and HBsAg secretion of HepG2.2.15 was significantly decreased by Gal-CSSO systems, and the maximum inhibition rates were 1.82-fold and 2.38-fold greater than those of commercial Lipofectamine 2000 (Lipo2000) systems. In vivo Gal-CSSO systems exhibited HC targeting and HC microenvironmental responsiveness without noticeable hepatotoxicity or systemic toxicity. The HBeAg and HBsAg titers of the HBV-infected mice were evidently decreased by Gal-CSSO systems, and the inhibition rates were 1.52-fold and 1.22-fold greater than those of Lipo2000 systems. This study presents a kind of glycolipid-like polymer micelles that promise efficient and safe gene therapy of HB.
Collapse
Affiliation(s)
- Jing Miao
- Department of Pharmacy, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Hangzhou 310003, China
| | - Xiqin Yang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Xuwei Shang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Zhe Gao
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qian Li
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yun Hong
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiaying Wu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Corresponding author: Jiaying Wu, PhD, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
- Corresponding author: Fuqiang Hu, PhD, College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
18
|
Perrone F, Craparo EF, Cemazar M, Kamensek U, Drago SE, Dapas B, Scaggiante B, Zanconati F, Bonazza D, Grassi M, Truong N, Pozzato G, Farra R, Cavallaro G, Grassi G. Targeted delivery of siRNAs against hepatocellular carcinoma-related genes by a galactosylated polyaspartamide copolymer. J Control Release 2021; 330:1132-1151. [PMID: 33212117 DOI: 10.1016/j.jconrel.2020.11.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/07/2020] [Accepted: 11/11/2020] [Indexed: 02/08/2023]
Abstract
Given the lack of effective treatments for Hepatocellular carcinoma (HCC), the development of novel therapeutic approaches is very urgent. Here, siRNAs were delivered to HCC cells by a synthetic polymer containing α,β-poly-(N-2-hydroxyethyl)-D,L-aspartamide-(PHEA) derivatized with diethylene triamine (DETA) and bearing in the side chain galactose (GAL) linked via a polyethylene glycol (PEG) to obtain (PHEA-DETA-PEG-GAL, PDPG). The GAL residue allows the targeting to the asialo-glycoprotein receptor (ASGPR), overexpressed in HCC cells compared to normal hepatocytes. Uptake studies performed using a model siRNA or a siRNA targeted against the enhanced green fluorescence protein, demonstrated the PDPG specific delivery of siRNA to HuH7 cells, a human cellular model of HCC. GAL-free copolymer (PHEA-DETA-PEG-NH2, PDP) or the chemical block of ASGPR, impaired PDPG targeting effectiveness in vitro. The specificity of PDPG delivery was confirmed in vivo in a mouse dorsal skinfold window chamber assay. Functional studies using siRNAs targeting the mRNAs of HCC-related genes (eEF1A1, eEF1A2 and E2F1) delivered by PDPG, significantly decreased HuH7 vitality/number and down regulated the expression of the target genes. Only minor effectiveness was in contrast observed for PDP. In IHH, a human model of normal hepatocytes with reduced ASGPR expression, PDPG barely reduced cell vitality. In a subcutaneous xenograft mouse model of HCC, PDPG-siRNAs reduced HCC tumor growth compared to controls without significant toxic effects. In conclusion, our study demonstrates the valuable potentials of PDPG for the specific delivery of siRNAs targeting HCC-related genes.
Collapse
Affiliation(s)
- Francesca Perrone
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, Trieste I-34149, Italy
| | - Emanuela Fabiola Craparo
- Department of Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, Palermo 90123, Italy
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, Ljubljana SI-1000, Slovenia; Faculty of Health Sciences, University of Primorska, Polje 42, SI-, Izola 6310, Slovenia
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, Ljubljana SI-1000, Slovenia
| | - Salvatore Emanuele Drago
- Department of Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, Palermo 90123, Italy
| | - Barbara Dapas
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, Trieste I-34149, Italy
| | - Bruna Scaggiante
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, Trieste I-34149, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, Trieste 447, Italy
| | - Debora Bonazza
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, Trieste 447, Italy
| | - Mario Grassi
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, Trieste I 34127, Italy
| | - Nhung Truong
- Stem Cell Research and Application Laboratory - VNUHCM - University of Science, Ho Chi Minh city, Viet Nam
| | - Gabriele Pozzato
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, Trieste 447, Italy
| | - Rossella Farra
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, Trieste I-34149, Italy.
| | - Gennara Cavallaro
- Department of Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, Palermo 90123, Italy.
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, Trieste I-34149, Italy; Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada di Fiume, Trieste 447, Italy
| |
Collapse
|
19
|
Bockman MR, Dalal RJ, Kumar R, Reineke TM. Facile synthesis of GalNAc monomers and block polycations for hepatocyte gene delivery. Polym Chem 2021. [DOI: 10.1039/d1py00250c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Here, we present a facile synthetic route for a monomer displaying N-acetyl-d-galactosamine and subsequent copolymerization in a block format with cationic subunits readily accessing liver-targeted polymeric pDNA delivery vehicles with low toxicity.
Collapse
Affiliation(s)
| | - Rishad J. Dalal
- Department of Chemistry
- University of Minnesota
- Minneapolis
- USA
| | - Ramya Kumar
- Department of Chemistry
- University of Minnesota
- Minneapolis
- USA
| | | |
Collapse
|
20
|
Pereira-Silva M, Jarak I, Santos AC, Veiga F, Figueiras A. Micelleplex-based nucleic acid therapeutics: From targeted stimuli-responsiveness to nanotoxicity and regulation. Eur J Pharm Sci 2020; 153:105461. [DOI: 10.1016/j.ejps.2020.105461] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022]
|
21
|
Zhang M, Weng Y, Cao Z, Guo S, Hu B, Lu M, Guo W, Yang T, Li C, Yang X, Huang Y. ROS-Activatable siRNA-Engineered Polyplex for NIR-Triggered Synergistic Cancer Treatment. ACS APPLIED MATERIALS & INTERFACES 2020; 12:32289-32300. [PMID: 32584027 DOI: 10.1021/acsami.0c06614] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Small interfering RNA (siRNA) shows excellent pharmaceutical prospects in treating diverse life-threatening diseases. Photodynamic therapy (PDT) is a clinically employed noninvasive treatment method that can trigger selective damage toward targeted tissue and cells. However, insufficient delivery of siRNA and photosensitizer to cancer cells remarkably hindered the application of siRNA and PDT in the treatment of cancer. In this study, a unique reactive oxygen species (ROS)-activatable polyplex, which consists of the PEGylated cationic polymer, ROS-cleavable linker, photosensitizer Ce6, and RRM2-against siRNA, termed PPTC/siRNA, was engineered. Upon irradiation of near-infrared (NIR) light, the polyplex efficiently generated ROS, which triggered degradation of the ROS-sensitive linker, disassembling the complex, destabilization of the cell membrane, and significantly accelerated cellular entry and endosomal escape of siRNA. Besides achieving effective siRNA internalization and gene silence in cancer cells in vitro, PPTC/siRNA synergistically inhibited tumor growth in both cell line-derived xenograft and patient-derived xenograft hepatocellular carcinoma murine models by repressing the RRM2 expression (reducing cell proliferation) and triggering photodynamic killing (enhancing cell apoptosis). The proposed polyplex also showed ideal safety profiles both in cell line and in animal. It provides a novel strategy for NIR-triggered RNAi and PDT combinational cancer treatment.
Collapse
Affiliation(s)
- Mengjie Zhang
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Institute of Engineering Medicine; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yuhua Weng
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Institute of Engineering Medicine; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Ziyang Cao
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Shuai Guo
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Institute of Engineering Medicine; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Bo Hu
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Institute of Engineering Medicine; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Mei Lu
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Institute of Engineering Medicine; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Weisheng Guo
- Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P. R. China
| | - Tongren Yang
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Institute of Engineering Medicine; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Chunhui Li
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Institute of Engineering Medicine; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Xianzhu Yang
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China
| | - Yuanyu Huang
- School of Life Science; Advanced Research Institute of Multidisciplinary Science; Institute of Engineering Medicine; Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, P. R. China
- School of Materials and the Environment, Beijing Institute of Technology, Zhuhai 519085, P. R. China
| |
Collapse
|
22
|
Peng Y, Bariwal J, Kumar V, Tan C, Mahato RI. Organic Nanocarriers for Delivery and Targeting of Therapeutic Agents for Cancer Treatment. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Peng
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Jitender Bariwal
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Virender Kumar
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug DeliveryUniversity of Mississippi University MS 38677 USA
| | - Ram I. Mahato
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| |
Collapse
|
23
|
Wang H, Liu Y, Xu M, Cheng J. Azido-galactose outperforms azido-mannose for metabolic labeling and targeting of hepatocellular carcinoma. Biomater Sci 2019; 7:4166-4173. [PMID: 31368459 DOI: 10.1039/c9bm00898e] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Metabolic glycoengineering of unnatural monosaccharides provides a facile method to label cancer cells with chemical tags for glycan imaging and cancer targeting. Multiple types of monosaccharides have been utilized for metabolic cell labeling. However, the comparison of different types of monosaccharides in labeling efficiency and selectivity has not been reported. In this study, we compared N-azidoacetylgalactosamine (GalAz) and N-azidoacetylmannosamine (ManAz) for metabolic labeling of HepG2 hepatocellular carcinoma in vitro and in vivo. GalAz showed higher labeling efficiency at low concentrations, and outperformed ManAz in metabolic labeling of HepG2 tumors in vivo. GalAz mediated labeling of HepG2 tumors with azido groups significantly improved the tumor accumulation of dibenzocyclooctyne (DBCO)-Cy5 and DBCO-doxorubicin conjugate via efficient Click chemistry. This study, for the first time, uncovered the distinct labeling efficiency and selectivity of different unnatural monosaccharides in liver cancers.
Collapse
Affiliation(s)
- Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Yang Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Ming Xu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| |
Collapse
|
24
|
Liang S, Zheng J, Wu W, Li Q, Saw PE, Chen J, Xu X, Yao H, Yao Y. A Robust Nanoparticle Platform for RNA Interference in Macrophages to Suppress Tumor Cell Migration. Front Pharmacol 2018; 9:1465. [PMID: 30618757 PMCID: PMC6302002 DOI: 10.3389/fphar.2018.01465] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 11/30/2018] [Indexed: 11/13/2022] Open
Abstract
Macrophages are one of the most abundant immune cells in the solid tumor and their increased density is associated with the specific pathological features of cancers, including invasiveness, metastasis, immunosuppression, neovascularization, and poor response to therapy. Therefore, reprogramming macrophage behavior is emerging as a promising therapeutic modality for cancer treatment. RNA interference (RNAi) technology is one of the powerful strategies for the regulation of macrophage activities by silencing specific genes. However, as polyanionic biomacromolecules, RNAi therapeutics such as small interfering RNA (siRNA) cannot readily cross cell membrane and thus specific delivery vehicles are required to facilitate the cytosolic siRNA delivery. Herein, we developed a robust nanoparticle (NP) platform for efficient siRNA delivery and gene silencing in macrophages. This NP platform is composed of biodegradable poly (ethylene glycol)-b-poly (𝜀-caprolactone) (PEG-b-PCL), poly (𝜀-caprolactone)-b-poly (2-aminoethyl ethylene phosphate) (PCL-b-PPEEA), and PCL homopolymer. We chose CC-chemokine ligand 18 (CCL-18) as a proof of concept therapeutic target and our results demonstrate that the CCL-18 silencing in macrophages can significantly inhibit the migration of breast cancer cells. The successful regulation of the macrophage behavior demonstrated herein shows great potential as an effective strategy for cancer therapy.
Collapse
Affiliation(s)
- Shi Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junmeng Zheng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Quan Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yandan Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
25
|
Polyester based nanovehicles for siRNA delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:1006-1015. [DOI: 10.1016/j.msec.2018.05.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/12/2018] [Accepted: 05/07/2018] [Indexed: 12/18/2022]
|
26
|
Fu H, Zhang X, Wang Q, Sun Y, Liu L, Huang L, Ding L, Shen M, Zhang L, Duan Y. Simple and rational design of a polymer nano-platform for high performance of HCV related miR-122 reduction in the liver. Biomater Sci 2018; 6:2667-2680. [PMID: 30209483 DOI: 10.1039/c8bm00639c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
microRNA-122 (miR-122) is a kind of non-coding RNA expressed specifically in the liver and accumulating evidence elucidates its relationship with HCV virus replication. The utilization of anti-miRNA oligonucleotide (antimiR) offers tremendous potential for future HCV infection therapy. However, multiple existing problems, such as targeting and stability, impede in vivo application of antimiR. To overcome them, we synthesized monomethoxy (polyethylene glycol)-poly (d,l lactide-co-glycolide)-poly (l-lysine) (mPEG-b-PLGA-b-PLL) materials to deliver miR-122 antagomir (AN), and formed stable and well-distributed AN-loaded mPEG-b-PLGA-b-PLL nanoparticles (NP-AN). NP-AN showed a high degree of miR-122 inhibition after 72 h in vitro. In vivo results showed an NP-AN "leak" through a hepatic sinusoid to reach hepatocytes and over 90% reduction of miR-122 after being injected with NP-AN for 72 h. Besides, the inhibition of miR-122 lasted for 28 days with limited dosage in vivo. This study strongly suggests that the silencing of miR-122 was enhanced and the reduction of miR-122 expression could be extended by utilizing an mPEG-b-PLGA-b-PLL nano-platform, which potentially facilitate further studies on miRNA function loss and related RNAi therapy for HCV infection.
Collapse
Affiliation(s)
- Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zhou L, Xi Y, Chen M, Niu W, Wang M, Ma PX, Lei B. A highly antibacterial polymeric hybrid micelle with efficiently targeted anticancer siRNA delivery and anti-infection in vitro/in vivo. NANOSCALE 2018; 10:17304-17317. [PMID: 30198034 DOI: 10.1039/c8nr03001d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Most of the diseases such as tumors are usually accompanied by microbial infection, especially after surgical operation, which prevents successful cancer therapy. It is necessary to develop a safe and efficient siRNA delivery vector with high anti-bacterial capability. Here, three multifunctional polymeric hybrid micelles (PHM1, PHM2 and PHM3) with high antimicrobial activity were prepared by mixing polymers PEG-b-P3/4HB-b-PEI-b-FA (EHP-FA) and PEG-b-P3/4HB-b-EPL (EHE) copolymer at different mixing ratios and evaluated for targeted siRNA delivery and anti-infection applications. The PHM micelles, taking advantage of the binding ability of EHE and the protection ability of EHP-FA, could effectively combine, protect siRNA, release complexed siRNA and target cancer cells. Additionally, PHM micelles displayed good hemocompatibility, lower cytotoxicity and higher gene silencing efficiency than commercial PEI (25 kDa) in A549, HeLa, HepG2 and C2C12 cells. Through optimizing the ratio of EHP-FA and EHE, PHM/sip65 showed a high p65 gene silencing efficiency above 90% in various cancer cells, which were significantly higher than EHP-FA/sip65 alone and EHE/sip65 complexes. Furthermore, PHM2 micelles showed excellent antimicrobial activity towards positive bacteria (S. aureus) in vitro and in vivo. Our study may provide a facile strategy to develop multifunctional polymer gene vectors for highly promising siRNA delivery and anti-infection.
Collapse
Affiliation(s)
- Li Zhou
- Frontier Institute of Science and Technology, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | | | | | | | | | | | | |
Collapse
|
28
|
Kuruvilla SP, Tiruchinapally G, Kaushal N, ElSayed ME. Effect of N-acetylgalactosamine ligand valency on targeting dendrimers to hepatic cancer cells. Int J Pharm 2018; 545:27-36. [DOI: 10.1016/j.ijpharm.2018.04.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 02/28/2018] [Accepted: 04/13/2018] [Indexed: 12/21/2022]
|
29
|
Dong H, Wu G, Xu H, Zhang C, Wang J, Gao M, Pang Y, Zhang H, Zhang B, Tian Y, Li Q. N-acetylaminogalactosyl-decorated biodegradable PLGA-TPGS copolymer nanoparticles containing emodin for the active targeting therapy of liver cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:260-272. [PMID: 29914275 DOI: 10.1080/21691401.2018.1455055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Primary liver cancer (PLC) is one of the most common malignant tumours and has the third highest mortality rate worldwide. An active liver-targeting drug delivery system via the asialoglycoprotein receptors expressed in the hepatic parenchyma cells of mammals has become a research focus for the treatment of PLC. N-acetylaminogalactosyl-poly(lactide-co-glycolide)-succinyl-D-α-tocopherol polyethylene glycol 1000 succinate (GalNAc-PLGA-sTPGS) was synthesized to achieve active liver-targeting properties. Emodin (EMO)-loaded GalNAc-PLGA-sTPGS nanoparticles (EGPTN) were prepared with EMO which was selected for its potential antitumour efficacy. The in vitro cellular uptake, mechanism, cytotoxicity, and apoptosis of HepG2 cells were analyzed. The in vivo therapeutic effects of EGPTN were assessed in a PLC mouse model. The results showed that GalNAc-PLGA-sTPGS was successfully synthesized. The cellular uptake assay demonstrated that coumarin 6-loaded GalNAc-PLGA-sTPGS nanoparticles had superior active liver-targeting properties. The results of the cytotoxity and apoptosis studies indicated that EGPTN achieved the highest levels of cytotoxicity and cell apoptotic rate among the nanoparticles examined. Furthermore, EGPTN showed better in vivo therapeutic effects and anticancer efficacy in the PLC mice than did the other groups. Therefore, EGPTN enhanced the anticancer effect of EMO both in vitro and in vivo, making it a potential option for the treatment of PLC.
Collapse
Affiliation(s)
- Hao Dong
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Guoyu Wu
- b Department of Pharmaceutics , the First Affiliated Hospital of Dalian Medical University , Dalian China
| | - Hong Xu
- c College of Basic Medical Sciences , Dalian Medical University , Dalian , China
| | - Chenghong Zhang
- c College of Basic Medical Sciences , Dalian Medical University , Dalian , China
| | - Jiao Wang
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Meng Gao
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Yue Pang
- d College of Life Science , Liaoning Normal University , Dalian , China
| | - Houli Zhang
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Baojing Zhang
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Yan Tian
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Qingwei Li
- d College of Life Science , Liaoning Normal University , Dalian , China
| |
Collapse
|
30
|
Wu S, Li N, Yang C, Yan L, Liang X, Ren M, Yang L. Synthesis of cationic branched tea polysaccharide derivatives for targeted delivery of siRNA to hepatocytes. Int J Biol Macromol 2018; 118:808-815. [PMID: 29857104 DOI: 10.1016/j.ijbiomac.2018.05.221] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/20/2018] [Accepted: 05/28/2018] [Indexed: 01/01/2023]
Abstract
The cationic branched tea polysaccharide (CTPSA) derivative bearing N-acylurea and 3-(dimethylamino)-1-propylamine residues was synthesized and characterized using FTIR and 1H NMR spectroscopy. A nonspecific siRNA (NsiRNA) was used as a model molecule of functional siRNA that could downregulate over-expressed glycometabolism enzymes in the liver. The result from the agarose gel electrophoresis confirmed that the CTPSA and NsiRNA could form stable complexes when their weight ratio was larger than 18. The zeta potentials and sizes of the complexes were in the range of +8-+15 mv and 120-150 nm, respectively. The CTPSA/NsiRNA complex was observed as nanoparticles with a spherical shape of approximately 100 nm using scanning electron microscopy. The CTPSA derivative and the CTPSA/NsiRNA complexes exhibited lower cytotoxicity in HL-7702 cells when compared with the branched PEI (bPEI) and bPEI/NsiRNA complexes assessed by the Cell Counting Kit-8 assay. The results of flow cytometric analysis and laser confocal microscopy indicated that the CTPSA derivative could effectively target the transfer of the NsiRNA to HL-7702 cells. This work provides a potential approach to promote the CTPSA derivative as a nonviral vector for targeted delivery of functional siRNA to hepatocytes.
Collapse
Affiliation(s)
- Shuyun Wu
- Department of Polymer and Material Science, School of Chemistry, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Provincial Key Laboratory for High Performance Polymer-based Composites, Sun Yat-Sen University, Guangzhou 510275, China
| | - Na Li
- Department of Endocrinology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Chuan Yang
- Department of Endocrinology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Li Yan
- Department of Endocrinology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xuan Liang
- Department of Polymer and Material Science, School of Chemistry, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Provincial Key Laboratory for High Performance Polymer-based Composites, Sun Yat-Sen University, Guangzhou 510275, China
| | - Meng Ren
- Department of Endocrinology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Liqun Yang
- Department of Polymer and Material Science, School of Chemistry, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Guangdong Provincial Key Laboratory for High Performance Polymer-based Composites, Sun Yat-Sen University, Guangzhou 510275, China.
| |
Collapse
|
31
|
Farra R, Musiani F, Perrone F, Čemažar M, Kamenšek U, Tonon F, Abrami M, Ručigaj A, Grassi M, Pozzato G, Bonazza D, Zanconati F, Forte G, El Boustani M, Scarabel L, Garziera M, Russo Spena C, De Stefano L, Salis B, Toffoli G, Rizzolio F, Grassi G, Dapas B. Polymer-Mediated Delivery of siRNAs to Hepatocellular Carcinoma: Variables Affecting Specificity and Effectiveness. Molecules 2018; 23:777. [PMID: 29597300 PMCID: PMC6017305 DOI: 10.3390/molecules23040777] [Citation(s) in RCA: 14] [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] [Received: 02/26/2018] [Revised: 03/22/2018] [Accepted: 03/23/2018] [Indexed: 02/06/2023] Open
Abstract
Despite the advances in anticancer therapies, their effectiveness for many human tumors is still far from being optimal. Significant improvements in treatment efficacy can come from the enhancement of drug specificity. This goal may be achieved by combining the use of therapeutic molecules with tumor specific effects and delivery carriers with tumor targeting ability. In this regard, nucleic acid-based drug (NABD) and particularly small interfering RNAs (siRNAs), are attractive molecules due to the possibility to be engineered to target specific tumor genes. On the other hand, polymeric-based delivery systems are emerging as versatile carriers to generate tumor-targeted delivery systems. Here we will focus on the most recent findings in the selection of siRNA/polymeric targeted delivery systems for hepatocellular carcinoma (HCC), a human tumor for which currently available therapeutic approaches are poorly effective. In addition, we will discuss the most attracting and, in our opinion, promising siRNA-polymer combinations for HCC in relation to the biological features of HCC tissue. Attention will be also put on the mathematical description of the mechanisms ruling siRNA-carrier delivery, this being an important aspect to improve effectiveness reducing the experimental work.
Collapse
Affiliation(s)
- Rossella Farra
- Department of Engineering and Architecture, University of Trieste, Via Alfonso Valerio, 6/A, I-34127 Trieste, Italy.
| | - Francesco Musiani
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, I-40127 Bologna, Italy.
| | - Francesca Perrone
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy.
| | - Maja Čemažar
- Department of Experimental Oncology, Institute of Oncology, Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia.
- Faculty of Health Sciences, University of Primorska, Polje 42, SI-6310 Izola, Slovenia.
| | - Urška Kamenšek
- Department of Experimental Oncology, Institute of Oncology, Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia.
| | - Federica Tonon
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy.
| | - Michela Abrami
- Department of Engineering and Architecture, University of Trieste, Via Alfonso Valerio, 6/A, I-34127 Trieste, Italy.
| | - Aleš Ručigaj
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia.
| | - Mario Grassi
- Department of Engineering and Architecture, University of Trieste, Via Alfonso Valerio, 6/A, I-34127 Trieste, Italy.
| | - Gabriele Pozzato
- Department of "Scienze Mediche, Chirurgiche e della Salute", University of Trieste, Cattinara Hospital, Strada di Fiume 447, I-34149 Trieste, Italy.
| | - Deborah Bonazza
- Department of "Scienze Mediche, Chirurgiche e della Salute", University of Trieste, Cattinara Hospital, Strada di Fiume 447, I-34149 Trieste, Italy.
| | - Fabrizio Zanconati
- Department of "Scienze Mediche, Chirurgiche e della Salute", University of Trieste, Cattinara Hospital, Strada di Fiume 447, I-34149 Trieste, Italy.
| | - Giancarlo Forte
- Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne's University Hospital, Studenstka 6, 656 91 Brno, Czech Republic.
| | - Maguie El Boustani
- Experimental and Clinical Pharmacology Unit, C.R.O.-National Cancer Institute, via Franco Gallini 2, I-33081 Aviano (PN), Italy.
- Doctoral School in Molecular Biomedicine, University of Trieste, 34100 Trieste, Italy.
| | - Lucia Scarabel
- C.R.O.-National Cancer Institute, via Franco Gallini 2, I-33081 Aviano (PN), Italy.
| | - Marica Garziera
- Experimental and Clinical Pharmacology Unit, C.R.O.-National Cancer Institute, via Franco Gallini 2, I-33081 Aviano (PN), Italy.
| | - Concetta Russo Spena
- Experimental and Clinical Pharmacology Unit, C.R.O.-National Cancer Institute, via Franco Gallini 2, I-33081 Aviano (PN), Italy.
- Doctoral School in Chemistry, University of Trieste, 34100 Trieste, Italy.
| | - Lucia De Stefano
- Experimental and Clinical Pharmacology Unit, C.R.O.-National Cancer Institute, via Franco Gallini 2, I-33081 Aviano (PN), Italy.
- Doctoral School in Chemistry, University of Trieste, 34100 Trieste, Italy.
| | - Barbara Salis
- Experimental and Clinical Pharmacology Unit, C.R.O.-National Cancer Institute, via Franco Gallini 2, I-33081 Aviano (PN), Italy.
- Doctoral School in Molecular Biomedicine, University of Trieste, 34100 Trieste, Italy.
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, C.R.O.-National Cancer Institute, via Franco Gallini 2, I-33081 Aviano (PN), Italy.
| | - Flavio Rizzolio
- Experimental and Clinical Pharmacology Unit, C.R.O.-National Cancer Institute, via Franco Gallini 2, I-33081 Aviano (PN), Italy.
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University, via Torino 155, I-30172 Mestre (Venezia), Italy.
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy.
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia.
| | - Barbara Dapas
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy.
| |
Collapse
|
32
|
Nikam RR, Gore KR. Journey of siRNA: Clinical Developments and Targeted Delivery. Nucleic Acid Ther 2018; 28:209-224. [PMID: 29584585 DOI: 10.1089/nat.2017.0715] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Since the evolutionary discovery of RNA interference and its utilization for gene knockdown in mammalian cell, a remarkable progress has been achieved in small interfering RNA (siRNA) therapeutics. siRNA is a promising tool, utilized as therapeutic agent against various diseases. Despite its significant potential benefits, safe, efficient, and target oriented delivery of siRNA is one of the major challenges in siRNA therapeutics. This review covers major achievements in clinical trials and targeted delivery of siRNAs using various targeting ligand-receptor pair. Local and systemically administered siRNA drug candidates at various phases in clinical trials are described in this review. This review also provides a deep insight in development of targeted delivery of siRNA. Various targeting ligand-siRNA pair with complexation and conjugation approaches are discussed in this review. This will help to achieve further optimization and development in targeted delivery of siRNAs to achieve higher gene silencing efficiency with lowest siRNA dose availability.
Collapse
Affiliation(s)
| | - Kiran R Gore
- Department of Chemistry, University of Mumbai , Mumbai, India
| |
Collapse
|
33
|
Mazur J, Roy K, Kanwar JR. Recent advances in nanomedicine and survivin targeting in brain cancers. Nanomedicine (Lond) 2017; 13:105-137. [PMID: 29161215 DOI: 10.2217/nnm-2017-0286] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Brain cancer is a highly lethal disease, especially devastating toward both the elderly and children. This cancer has no therapeutics available to combat it, predominately due to the blood-brain barrier (BBB) preventing treatments from maintaining therapeutic levels within the brain. Recently, nanoparticle technology has entered the forefront of cancer therapy due to its ability to deliver therapeutic effects while potentially passing physiological barriers. Key nanoparticles for brain cancer treatment include glutathione targeted PEGylated liposomes, gold nanoparticles, superparamagnetic iron oxide nanoparticles and nanoparticle-albumin bound drugs, with these being discussed throughout this review. Recently, the survivin protein has gained attention as it is over-expressed in a majority of tumors. This review will briefly discuss the properties of survivin, while focusing on how both nanoparticles and survivin-targeting treatments hold potential as brain cancer therapies. This review may provide useful insight into new brain cancer treatment options, particularly survivin inhibition and nanomedicine.
Collapse
Affiliation(s)
- Jake Mazur
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| | - Kislay Roy
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| | - Jagat R Kanwar
- Nanomedicine-Laboratory of Immunology & Molecular Biomedical Research, Centre for Molecular and Medical Research (CMMR), School of Medicine, Faculty of Health, Deakin University, Waurn Ponds, Geelong VIC 3217, Australia
| |
Collapse
|
34
|
Cavallaro G, Farra R, Craparo EF, Sardo C, Porsio B, Giammona G, Perrone F, Grassi M, Pozzato G, Grassi G, Dapas B. Galactosylated polyaspartamide copolymers for siRNA targeted delivery to hepatocellular carcinoma cells. Int J Pharm 2017; 525:397-406. [PMID: 28119125 DOI: 10.1016/j.ijpharm.2017.01.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 02/07/2023]
Abstract
The limited efficacy of available treatments for hepatocellular carcinoma (HCC) requires the development of novel therapeutic approaches. We synthesized a novel cationic polymer based on α,β-poly-(N-2-hydroxyethyl)-d,L-aspartamide (PHEA) for drug delivery to HCC cells. The copolymer was synthesized by subsequent derivatization of PHEA with diethylene triamine (DETA) and with a polyethylene glycol (PEG) derivative bearing galactose (GAL) molecules, obtaining the cationic derivative PHEA-DETA-PEG-GAL. PHEA-DETA-PEG-GAL has suitable chemical-physical characteristics for a potential systemic use and can effectively deliver a siRNA (siE2F1) targeted against the transcription factor E2F1, a gene product involved in HCC. The presence of GAL residues in the polyplexes allows the targeting of HCC cells that express the asialo-glycoprotein receptor (ASGP-R). In these cells, but not in ASGP-R non-expressing cells, PHEA-DETA-PEG-GAL/siE2F1 polyplexes induce the reduction of the mRNA and protein levels of E2F1 and of E2F1-regulated genes, all involved in the promotion of the G1/S phase transition. This results in a decrease of cell proliferation with a G1/G0 phase cells accumulation. Notably, removal of GAL residue almost completely abrogates the targeting capacity of the developed polyplexes. In conclusion, the generated polyplexes demonstrate the potential to effectively contributing to the development of novel anti-HCC therapeutic approaches via a siRNA-targeted delivery.
Collapse
Affiliation(s)
- Gennara Cavallaro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Rossella Farra
- Department of Industrial Engineering and Information Technology, University of Trieste, Italy
| | - Emanuela Fabiola Craparo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Carla Sardo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Barbara Porsio
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Gaetano Giammona
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche, Farmaceutiche (STEBICEF), Lab of Biocompatible Polymers, University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | | | - Mario Grassi
- Department of Industrial Engineering and Information Technology, University of Trieste, Italy
| | - Gabriele Pozzato
- Department of 'Medical, Surgery and Health Sciences, University of Trieste, Cattinara Hospital, Italy
| | | | - Barbara Dapas
- Department of Life Sciences, University of Trieste, Italy
| |
Collapse
|
35
|
Scarabel L, Perrone F, Garziera M, Farra R, Grassi M, Musiani F, Russo Spena C, Salis B, De Stefano L, Toffoli G, Rizzolio F, Tonon F, Abrami M, Chiarappa G, Pozzato G, Forte G, Grassi G, Dapas B. Strategies to optimize siRNA delivery to hepatocellular carcinoma cells. Expert Opin Drug Deliv 2017; 14:797-810. [PMID: 28266887 DOI: 10.1080/17425247.2017.1292247] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/03/2017] [Indexed: 02/08/2023]
Abstract
hepatocellular carcinoma (hcc) is the predominant form of primary liver cancer and the second leading cause of cancer-associated mortality worldwide. available therapies for hcc have limited efficacy due to often late diagnosis and the general resistance of hcc to anti-cancer agents; therefore, the development of novel therapeutics is urgently required. small-interfering rna (sirna) molecules are short, double-stranded rnas that specifically recognize and bind the mrna of a target gene to inhibit gene expression. despite the great therapeutic potential of sirnas towards many human tumors including hcc, their use is limited by suboptimal delivery. Areas covered: In this review, we outline the current data regarding the therapeutic potential of siRNAs in HCC and describe the development of effective siRNA delivery systems. We detail the key problems associated with siRNA delivery and discuss the possible solutions. Finally, we provide examples of the various siRNA delivery strategies that have been employed in animal models of HCC and in human patients enrolled in clinical trials. Expert opinion: Despite the existing difficulties in siRNA delivery for HCC, the increasing scientific attention and breakthrough studies in this field is facilitating the design of novel and efficient technical solutions that may soon find practical applications.
Collapse
Affiliation(s)
- Lucia Scarabel
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Francesca Perrone
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
| | - Marica Garziera
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Rossella Farra
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Mario Grassi
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Francesco Musiani
- d Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Concetta Russo Spena
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Barbara Salis
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Lucia De Stefano
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Giuseppe Toffoli
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Flavio Rizzolio
- a Experimental and Clinical Pharmacology Unit , C.R.O. National Cancer Institute , Aviano , Italy
| | - Federica Tonon
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Michela Abrami
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
| | - Gianluca Chiarappa
- c Department of Engineering and Architecture , University of Trieste , Trieste , Italy
| | - Gabriele Pozzato
- e Department of 'Scienze Mediche, Chirurgiche e della Salute' , Cattinara University Hospital, University of Trieste , Trieste , Italy
| | - Giancarlo Forte
- f Center for Translational Medicine, International Clinical Research Center , St. Anne's University Hospital , Brno , Czech Republic
| | - Gabriele Grassi
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
- e Department of 'Scienze Mediche, Chirurgiche e della Salute' , Cattinara University Hospital, University of Trieste , Trieste , Italy
| | - Barbara Dapas
- b Department of Life Sciences, Cattinara University Hospital , University of Trieste , Trieste , Italy
| |
Collapse
|
36
|
Wu J, Qu W, Williford JM, Ren Y, Jiang X, Jiang X, Pan D, Mao HQ, Luijten E. Improved siRNA delivery efficiency via solvent-induced condensation of micellar nanoparticles. NANOTECHNOLOGY 2017; 28:204002. [PMID: 28266928 PMCID: PMC5790992 DOI: 10.1088/1361-6528/aa6519] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Efficient delivery of short interfering RNA (siRNA) remains one of the primary challenges of RNA interference therapy. Polyethylene glycol (PEG)ylated polycationic carriers have been widely used for the condensation of DNA and RNA molecules into complex-core micelles. The PEG corona of such nanoparticles can significantly improve their colloidal stability in serum, but PEGylation of the carriers also reduces their condensation capacity, hindering the generation of micellar particles with sufficient complex stability. This presents a particularly significant challenge for packaging siRNA into complex micelles, as it has a much smaller size and more rigid chain structure than DNA plasmids. Here, we report a new method to enhance the condensation of siRNA with PEGylated linear polyethylenimine using organic solvent and to prepare smaller siRNA nanoparticles with a more extended PEG corona and consequently higher stability. As a proof of principle, we have demonstrated the improved gene knockdown efficiency resulting from the reduced siRNA micelle size in mice livers following intravenous administration.
Collapse
Affiliation(s)
- Juan Wu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Wei Qu
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208
| | - John-Michael Williford
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - Yong Ren
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - Xuesong Jiang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218
- Translational Tissue Engineering Center and Whitaker Biomedical Engineering Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21287
| | - Xuan Jiang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Deng Pan
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland 21205
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
- Translational Tissue Engineering Center and Whitaker Biomedical Engineering Institute, Johns Hopkins School of Medicine, Baltimore, Maryland 21287
| | - Erik Luijten
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
37
|
Lou S, Zhang X, Zhang M, Ji S, Wang W, Zhang J, Li C, Kong D. Preparation of a dual cored hepatoma-specific star glycopolymer nanogel via arm-first ATRP approach. Int J Nanomedicine 2017; 12:3653-3664. [PMID: 28553105 PMCID: PMC5439995 DOI: 10.2147/ijn.s134367] [Citation(s) in RCA: 16] [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: 12/04/2022] Open
Abstract
A reductase-cleavable and thermo-responsive star-shaped polymer nanogel was prepared via an "arm-first" atom transfer radical polymerization approach. The nanogel consists of a thermo- and redox-sensitive core and a zwitterionic copolymer block. The dual sensitive core is composed of poly(N-isopropylacrylamide) that is formed by disulfide crosslinking of N-isopropylacrylamide. The zwitterionic copolymer block contains a poly(sulfobetaine methacrylate) component, a known anti-adsorptive moiety that extends blood circulation time, and a lactose motif of poly(2-lactobionamidoethyl methacrylamide) that specifically targets the asialoglycoprotein receptors (ASGP-Rs) of hepatoma. Doxorubicin (DOX) was encapsulated into the cross-linked nanogels via solvent extraction/evaporation method and dialysis; average diameter of both blank and DOX-loaded nanogels was ~120 nm. The multi-responsiveness of nanogel drug release in different temperatures and redox conditions was assessed. After 24 h, DOX release was only ~20% at 30°C with 0 mM glutathione (GSH), whereas over 90% DOX release was observed at 40°C and 10 mM GSH, evidence of dual responsiveness to temperature and reductase GSH. The IC50 value of DOX-loaded nanogels was much lower in human hepatoma (HepG2) cells compared to non-hepatic HeLa cells. Remarkably, DOX uptake of HepG2 cells differed substantially in the presence and absence of galactose (0.31 vs 1.42 µg/mL after 48 h of incubation). The difference was non-detectable in HeLa cells (1.21 vs 1.57 µg/mL after 48 h of incubation), indicating that the overexpression of ASGP-Rs leads to the DOX-loaded lactosylated nanogels actively targeting hepatoma. Our data indicate that the lactose-decorated star-shaped nanogels are dual responsive and hepatoma targeted, and could be employed as hepatoma-specific anti-cancer drug delivery vehicle for cancer chemotherapy.
Collapse
Affiliation(s)
- Shaofeng Lou
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University
| | - Xiuyuan Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science, Tianjin, People’s Republic of China
| | - Mingming Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science, Tianjin, People’s Republic of China
| | - Shenglu Ji
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science, Tianjin, People’s Republic of China
| | - Ju Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University
| | - Chen Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science, Tianjin, People’s Republic of China
| | - Deling Kong
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science, Tianjin, People’s Republic of China
| |
Collapse
|
38
|
Greco CT, Muir VG, Epps TH, Sullivan MO. Efficient tuning of siRNA dose response by combining mixed polymer nanocarriers with simple kinetic modeling. Acta Biomater 2017; 50:407-416. [PMID: 28063990 PMCID: PMC5317101 DOI: 10.1016/j.actbio.2017.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 12/01/2016] [Accepted: 01/03/2017] [Indexed: 12/12/2022]
Abstract
Two of the most prominent challenges that limit the clinical success of siRNA therapies are a lack of control over cargo release from the delivery vehicle and an incomplete understanding of the link between gene silencing dynamics and siRNA dosing. Herein, we address these challenges through the formulation of siRNA polyplexes containing light-responsive polymer mixtures, whose varied compositions and triggered release behavior provide enhanced gene silencing and controlled dose responses that can be predicted by simple kinetic models. Through the straightforward mixing of two block copolymers, the level of gene knockdown was easily optimized to achieve the maximum level of GAPDH protein silencing in NIH/3T3 cells (~70%) using a single siRNA dose. The kinetic model was used to describe the dynamic changes in mRNA and protein concentrations in response to siRNA treatment. These predictions enabled the application of a second dose of siRNA to maximally suppress gene expression over multiple days, leading to a further 50% reduction in protein levels relative to those measured following a single dose. Furthermore, polyplexes remained dormant in cells until exposed to the photo-stimulus, demonstrating the complete control over siRNA activity as well as the stability of the nanocarriers. Thus, this work demonstrates that pairing advances in biomaterials design with simple kinetic modeling provides new insight into gene silencing dynamics and presents a powerful strategy to control gene expression through siRNA delivery. STATEMENT OF SIGNIFICANCE Our manuscript describes two noteworthy impacts: (1) we designed mixed polymer formulations to enhance gene silencing, and (2) we simultaneously developed a simple kinetic model for determining optimal siRNA dose responses to maintain silencing over several days. These advances address critical challenges in siRNA delivery and provide new opportunities in therapeutics development. The structure-function relationships prevalent in these formulations were established to enable tuning and forecasting of nanocarrier efficiency a priori, leading to siRNA dosing regimens able to maximally suppress gene expression. Our advances are significant because the mixed polymer formulations provide a straightforward and scalable approach to tailor siRNA delivery regimens. Moreover, the implementation of accurate dosing frameworks addresses a major knowledge gap that has hindered clinical implementation of siRNA.
Collapse
Affiliation(s)
- Chad T Greco
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Victoria G Muir
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Thomas H Epps
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA; Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
39
|
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
|
40
|
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: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
41
|
Yilmaz ZE, Jérôme C. Polyphosphoesters: New Trends in Synthesis and Drug Delivery Applications. Macromol Biosci 2016; 16:1745-1761. [PMID: 27654308 DOI: 10.1002/mabi.201600269] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/18/2016] [Indexed: 11/06/2022]
Abstract
Polymers with repeating phosphoester linkages in the backbone are biodegradable materials that emerge as a promising class of novel biomaterials, especially in the field of drug delivery systems. In contrast to aliphatic polyesters, the pentavalency of the phosphorus atom offers a large diversity of structures and as a consequence a wide range of properties for these materials. In this paper, it is focused on the synthesis of well-defined polyphosphoesters (PPEs) by organocatalyzed ring-opening polymerization, improving the functionalities by combination with click reactions, degradation of functional PPEs and their cytotoxicity, and inputs for applications in drug delivery.
Collapse
Affiliation(s)
- Zeynep Ergul Yilmaz
- Center for Education and Research on Macromolecules (CERM), University of Liège (ULg), CESAM-RU, Sart Tilman, Building B6a, Liège, B-4000, Belgium
| | - Christine Jérôme
- Center for Education and Research on Macromolecules (CERM), University of Liège (ULg), CESAM-RU, Sart Tilman, Building B6a, Liège, B-4000, Belgium
| |
Collapse
|
42
|
Cooper BM, Putnam D. Polymers for siRNA Delivery: A Critical Assessment of Current Technology Prospects for Clinical Application. ACS Biomater Sci Eng 2016; 2:1837-1850. [PMID: 33440520 DOI: 10.1021/acsbiomaterials.6b00363] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The number of polymer-based vectors for siRNA delivery in clinical trials lags behind other delivery strategies; however, the molecular architectures and chemical compositions available to polymers make them attractive candidates for further exploration. Polymer vectors are extensively investigated in academic laboratories worldwide with fundamental progress having recently been made in the areas of high-throughput screening, synthetic methods, cellular internalization, endosomal escape and computational prediction and analysis. This review assesses recent advances within the field and highlights relevant developments from within the complementary fields of nanotechnology and protein chemistry with the intent to propose future work that addresses key gaps within the current body of knowledge, potentially advancing the development of the next generation of polymeric vectors.
Collapse
Affiliation(s)
- Bailey M Cooper
- Meinig School of Biomedical Engineering and ‡Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - David Putnam
- Meinig School of Biomedical Engineering and Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
43
|
|
44
|
Pereira P, Barreira M, Queiroz JA, Veiga F, Sousa F, Figueiras A. Smart micelleplexes as a new therapeutic approach for RNA delivery. Expert Opin Drug Deliv 2016; 14:353-371. [DOI: 10.1080/17425247.2016.1214567] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
45
|
Kuai R, Li D, Chen YE, Moon JJ, Schwendeman A. High-Density Lipoproteins: Nature's Multifunctional Nanoparticles. ACS NANO 2016; 10:3015-41. [PMID: 26889958 PMCID: PMC4918468 DOI: 10.1021/acsnano.5b07522] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
High-density lipoproteins (HDL) are endogenous nanoparticles involved in the transport and metabolism of cholesterol, phospholipids, and triglycerides. HDL is well-known as the "good" cholesterol because it not only removes excess cholesterol from atherosclerotic plaques but also has anti-inflammatory and antioxidative properties, which protect the cardiovascular system. Circulating HDL also transports endogenous proteins, vitamins, hormones, and microRNA to various organs. Compared with other synthetic nanocarriers, such as liposomes, micelles, and inorganic and polymeric nanoparticles, HDL has unique features that allow them to deliver cargo to specific targets more efficiently. These attributes include their ultrasmall size (8-12 nm in diameter), high tolerability in humans (up to 8 g of protein per infusion), long circulating half-life (12-24 h), and intrinsic targeting properties to different recipient cells. Various recombinant ApoA proteins and ApoA mimetic peptides have been recently developed for the preparation of reconstituted HDL that exhibits properties similar to those of endogenous HDL and has a potential for industrial scale-up. In this review, we will summarize (a) clinical pharmacokinetics and safety of reconstituted HDL products, (b) comparison of HDL with inorganic and other organic nanoparticles,
Collapse
Affiliation(s)
- Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dan Li
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Y. Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, 1150 W Medical Center Dr, Ann Arbor, MI 48109, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence should be addressed to A. S. () or J.J.M. ()
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence should be addressed to A. S. () or J.J.M. ()
| |
Collapse
|
46
|
Dhande YK, Wagh BS, Hall BC, Sprouse D, Hackett PB, Reineke TM. N-Acetylgalactosamine Block-co-Polycations Form Stable Polyplexes with Plasmids and Promote Liver-Targeted Delivery. Biomacromolecules 2016; 17:830-40. [DOI: 10.1021/acs.biomac.5b01555] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yogesh K. Dhande
- Department of Chemical Engineering and Materials Science, and Center
for Genome Engineering, ‡Department of Chemistry and Center for Genome Engineering, and §Department of Genetics,
Cell Biology and Development, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Bharat S. Wagh
- Department of Chemical Engineering and Materials Science, and Center
for Genome Engineering, ‡Department of Chemistry and Center for Genome Engineering, and §Department of Genetics,
Cell Biology and Development, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Bryan C. Hall
- Department of Chemical Engineering and Materials Science, and Center
for Genome Engineering, ‡Department of Chemistry and Center for Genome Engineering, and §Department of Genetics,
Cell Biology and Development, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Dustin Sprouse
- Department of Chemical Engineering and Materials Science, and Center
for Genome Engineering, ‡Department of Chemistry and Center for Genome Engineering, and §Department of Genetics,
Cell Biology and Development, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Perry B. Hackett
- Department of Chemical Engineering and Materials Science, and Center
for Genome Engineering, ‡Department of Chemistry and Center for Genome Engineering, and §Department of Genetics,
Cell Biology and Development, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M. Reineke
- Department of Chemical Engineering and Materials Science, and Center
for Genome Engineering, ‡Department of Chemistry and Center for Genome Engineering, and §Department of Genetics,
Cell Biology and Development, and Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
47
|
Zuo ZQ, Chen KG, Yu XY, Zhao G, Shen S, Cao ZT, Luo YL, Wang YC, Wang J. Promoting tumor penetration of nanoparticles for cancer stem cell therapy by TGF-β signaling pathway inhibition. Biomaterials 2015; 82:48-59. [PMID: 26751819 DOI: 10.1016/j.biomaterials.2015.12.014] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 12/13/2015] [Indexed: 02/06/2023]
Abstract
Cancer stem cells (CSCs), which hold a high capacity for self-renewal, play a central role in the development, metastasis, and recurrence of various malignancies. CSCs must be eradicated to cure instances of cancer; however, because they can reside far from tumor vessels, they are not easily targeted by drug agents carried by nanoparticle-based drug delivery systems. We herein demonstrate that promoting tumor penetration of nanoparticles by transforming growth factor β (TGF-β) signaling pathway inhibition facilitates CSC therapy. In our study, we observed that although nanoparticles carrying siRNA targeting the oncogene polo-like kinase 1 (Plk1) efficiently killed breast CSCs derived from MDA-MB-231 cells in vitro, this intervention enriched CSCs in the residual tumor tissue following systemic treatment. However, inhibition of the TGF-β signaling pathway with LY364947, an inhibitor of TGF-β type I receptor, promoted the penetration of nanoparticles in tumor tissue, significantly ameliorating the intratumoral distribution of nanoparticles in MDA-MB-231 xenografts and further leading to enhanced internalization of nanoparticles by CSCs. As a result, synergistic treatment with a nanoparticle drug delivery system and LY364947 inhibited tumor growth and reduced the proportion of CSCs in vivo. This study suggests that enhanced tumor penetration of drug-carrying nanoparticles can enhance CSCs clearance in vivo and consequently provide superior anti-tumor effects.
Collapse
Affiliation(s)
- Zu-Qi Zuo
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, 230027, PR China
| | - Kai-Ge Chen
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, 230027, PR China
| | - Xiao-Yuan Yu
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, 230027, PR China
| | - Gui Zhao
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, 230027, PR China
| | - Song Shen
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, 230027, PR China
| | - Zhi-Ting Cao
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Anhui, 230027, PR China
| | - Ying-Li Luo
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, 230027, PR China
| | - Yu-Cai Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, 230027, PR China
| | - Jun Wang
- The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, 230027, PR China; Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Anhui, 230027, PR China; CAS Center for Excellence in Nanoscience, University of Science and Technology of China, Hefei, Anhui, 230026, PR China; Innovation Center for Cell Signaling Network, University of Science and Technology of China, Hefei, Anhui, 230027, PR China.
| |
Collapse
|
48
|
Mondal G, Kumar V, Shukla SK, Singh PK, Mahato RI. EGFR-Targeted Polymeric Mixed Micelles Carrying Gemcitabine for Treating Pancreatic Cancer. Biomacromolecules 2015; 17:301-13. [PMID: 26626700 DOI: 10.1021/acs.biomac.5b01419] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The objective of this study was to design GE11 peptide (YHWYGYTPQNVI) linked micelles of poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylene carbonate-graft-gemcitabine-graft-dodecanol (PEG-b-PCC-g-GEM-g-DC) for enhanced stability and target specificity of gemcitabine (GEM) to EGFR-positive pancreatic cancer cells. GE11-PEG-PCD/mPEG-b-PCC-g-GEM-g-DC mixed micelles showed EGFR-dependent enhanced cellular uptake, and cytotoxicity as compared to scrambled peptide HW12-PEG-PCD/mPEG-b-PCC-g-GEM-g-DC mixed micelles and unmodified mPEG-b-PCC-g-GEM-g-DC micelles. Importantly, GE11-linked mixed micelles preferentially accumulated in orthotopic pancreatic tumor and tumor vasculature at 24 h post systemic administration. GE11-linked mixed micelles inhibited orthotopic pancreatic tumor growth compared to HW12-linked mixed micelles, unmodified mPEG-b-PCC-g-GEM-g-DC micelles, and free GEM formulations. Tumor growth inhibition was mediated by apoptosis of tumor cells and endothelial cells as determined by immunohistochemical staining. In summary, GE11-linked mixed micelles is a promising approach to treat EGFR overexpressing cancers.
Collapse
Affiliation(s)
- Goutam Mondal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Surendra K Shukla
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center , Omaha, Nebraska United States
| | - Pankaj K Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center , Omaha, Nebraska United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| |
Collapse
|
49
|
Li H, Fu Y, Zhang T, Li Y, Hong X, Jiang J, Gong T, Zhang Z, Sun X. Rational Design of Polymeric Hybrid Micelles with Highly Tunable Properties to Co‐Deliver MicroRNA‐34a and Vismodegib for Melanoma Therapy. ADVANCED FUNCTIONAL MATERIALS 2015; 25:7457-7469. [DOI: 10.1002/adfm.201503115] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
A polymeric hybrid micelle (PHM) system with highly tunable properties is reported to co‐deliver small molecule and nucleic acid drugs for cancer therapy; this system is structurally simple and easy‐to‐fabricate. The PHM consists of two amphiphilic diblock copolymers, polycaprolactone‐polyethylenimine (PCL‐PEI) and polycaprolactone‐polyethyleneglycol (PCL‐PEG). PHMs are rationally designed with different physicochemical properties by simply adjusting the ratio of the two diblock copolymers and the near neutral PHM‐2 containing a low ratio of PCL‐PEI achieves the optimal balance between high tumor distribution and subsequent cellular uptake after intravenous injection. Encapsulating Hedgehog (Hh) pathway inhibitor vismodegib (VIS) and microRNA‐34a (miR‐34a) into PHM‐2 generates the VIS/PHM‐2/34a co‐delivery system. VIS/PHM‐2/34a shows synergistic anticancer efficacy in murine B16F10‐CD44+ cells, a highly metastatic tumor model of melanoma. VIS/PHM‐2/34a synergistically attenuates the expression of CD44, a vital receptor indicating the metastasis of melanoma. Intriguingly, inhibiting Hh pathway by VIS is accompanied by downregulation of CD44 expression, revealing that Hh signaling might be an upstream regulator of CD44 expression in melanoma. Thus, co‐delivery of miR‐34a and VIS demonstrates great potential in cancer therapy, and PHM offers a structurally simple and highly tunable platform for the co‐delivery of small molecule and nucleic acid drugs in tumor combination therapy.
Collapse
Affiliation(s)
- Hanmei Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education West China School of Pharmacy Sichuan University Chengdu 610041 P.R. China
| | - Yao Fu
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education West China School of Pharmacy Sichuan University Chengdu 610041 P.R. China
| | - Ting Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education West China School of Pharmacy Sichuan University Chengdu 610041 P.R. China
| | - Yanping Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education West China School of Pharmacy Sichuan University Chengdu 610041 P.R. China
| | - Xiaoyu Hong
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education West China School of Pharmacy Sichuan University Chengdu 610041 P.R. China
| | - Jiayu Jiang
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education West China School of Pharmacy Sichuan University Chengdu 610041 P.R. China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education West China School of Pharmacy Sichuan University Chengdu 610041 P.R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education West China School of Pharmacy Sichuan University Chengdu 610041 P.R. China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems Ministry of Education West China School of Pharmacy Sichuan University Chengdu 610041 P.R. China
| |
Collapse
|
50
|
|