1
|
Yin M, Liu L, Yan Y, Wang H, Li W, Dong Y, Kong G. A targeting nanoplatform for chemo-photothermal synergistic therapy of small-cell lung cancer. Int J Cancer 2024; 155:2094-2106. [PMID: 38985144 DOI: 10.1002/ijc.35065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 07/11/2024]
Abstract
The precise delivery of drugs to tumor sites and the thermoresistance of tumors remain major challenges in photothermal therapy (PTT). Somatostatin receptor 2 (SSTR2) is proposed as an ideal target for the precise treatment of SCLC. We developed a targeting nano-drug delivery system comprising anti-SSTR2 monoclonal antibody (MAb) surface-modified nanoparticles co-encapsulating Cypate and gambogic acid (GA). The formed SGCPNs demonstrated excellent monodispersity, physiological stability, preferable biocompatibility, and resultant efficient photothermal conversion efficacy. SGCPNs were quickly internalized by SSTR2-overexpressing SCLC cells, triggering the release of GA under acidic and near-infrared (NIR) laser irradiation environments, leading to their escape from lysosomes to the cytosol and then diffusion into the nucleus. SGCPNs can not only decrease the cell survival rate but also inhibit the activity of heat shock protein 90 (HSP90). SGCPNs can be precisely delivered to xenograft tumors of SSTR2-positive SCLC in vivo. Upon NIR laser irradiation, therapy of SGCPNs showed significant tumor regression. In conclusion, SGCPNs provide a new chemo-photothermal synergistic treatment strategy for targeting SCLC.
Collapse
Affiliation(s)
- Moli Yin
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
| | - Lei Liu
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
| | - Yu Yan
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Huiyan Wang
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
| | - Wenliang Li
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
| | - Yuan Dong
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Guangyao Kong
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
2
|
Pérez-Rubio P, Vendrell-Flotats M, Romero EL, Enemark-Rasmussen K, Cervera L, Gòdia F, Lavado-García J. Internalization of PEI-based complexes in transient transfection of HEK293 cells is triggered by coalescence of membrane heparan sulfate proteoglycans like Glypican-4. Biomed Pharmacother 2024; 176:116893. [PMID: 38850653 DOI: 10.1016/j.biopha.2024.116893] [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: 03/22/2024] [Revised: 05/23/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024] Open
Abstract
Polymer-cationic mediated gene delivery is a well-stablished strategy of transient gene expression (TGE) in mammalian cell cultures. Nonetheless, its industrial implementation is hindered by the phenomenon known as cell density effect (CDE) that limits the cell density at which cultures can be efficiently transfected. The rise in personalized medicine and multiple cell and gene therapy approaches based on TGE, make more relevant to understand how to circumvent the CDE. A rational study upon DNA/PEI complex formation, stability and delivery during transfection of HEK293 cell cultures has been conducted, providing insights on the mechanisms for polyplexes uptake at low cell density and disruption at high cell density. DNA/PEI polyplexes were physiochemically characterized by coupling X-ray spectroscopy, confocal microscopy, cryo-transmission electron microscopy (TEM) and nuclear magnetic resonance (NMR). Our results showed that the ionic strength of polyplexes significantly increased upon their addition to exhausted media. This was reverted by depleting extracellular vesicles (EVs) from the media. The increase in ionic strength led to polyplex aggregation and prevented efficient cell transfection which could be counterbalanced by implementing a simple media replacement (MR) step before transfection. Inhibiting and labeling specific cell-surface proteoglycans (PGs) species revealed different roles of PGs in polyplexes uptake. Importantly, the polyplexes uptake process seemed to be triggered by a coalescence phenomenon of HSPG like glypican-4 around polyplex entry points. Ultimately, this study provides new insights into PEI-based cell transfection methodologies, enabling to enhance transient transfection and mitigate the cell density effect (CDE).
Collapse
Affiliation(s)
- Pol Pérez-Rubio
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - Meritxell Vendrell-Flotats
- Servei de Microscòpia, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - Elianet Lorenzo Romero
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | | | - Laura Cervera
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain; Serra Hunter Lecturer Professor.
| | - Francesc Gòdia
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - Jesús Lavado-García
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby Kgs 2800, Denmark.
| |
Collapse
|
3
|
Delgado Gonzalez B, Lopez-Blanco R, Parcero-Bouzas S, Barreiro-Piñeiro N, Garcia-Abuin L, Fernandez-Megia E. Dynamic Covalent Boronate Chemistry Accelerates the Screening of Polymeric Gene Delivery Vectors via In Situ Complexation of Nucleic Acids. J Am Chem Soc 2024; 146:17211-17219. [PMID: 38864331 PMCID: PMC11212051 DOI: 10.1021/jacs.4c03384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
Gene therapy provides exciting new therapeutic opportunities beyond the reach of traditional treatments. Despite the tremendous progress of viral vectors, their high cost, complex manufacturing, and side effects have encouraged the development of nonviral alternatives, including cationic polymers. However, these are less efficient in overcoming cellular barriers, resulting in lower transfection efficiencies. Although the exquisite structural tunability of polymers might be envisaged as a versatile tool for improving transfection, the need to fine-tune several structural parameters represents a bottleneck in current screening technologies. By taking advantage of the fast-forming and strong boronate ester bond, an archetypal example of dynamic covalent chemistry, a highly adaptable gene delivery platform is presented, in which the polycation synthesis and pDNA complexation occur in situ. The robustness of the strategy entitles the simultaneous evaluation of several structural parameters at will, enabling the accelerated screening and adaptive optimization of lead polymeric vectors using dynamic covalent libraries.
Collapse
Affiliation(s)
- Bruno Delgado Gonzalez
- Centro
Singular de Investigación en Química Biolóxica
e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| | - Roi Lopez-Blanco
- Centro
Singular de Investigación en Química Biolóxica
e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| | - Samuel Parcero-Bouzas
- Centro
Singular de Investigación en Química Biolóxica
e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| | - Natalia Barreiro-Piñeiro
- Centro
Singular de Investigación en Química Biolóxica
e Materiais Moleculares (CIQUS), Departamento de Bioquímica
e Bioloxía Molecular, Universidade
de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| | - Lucas Garcia-Abuin
- Centro
Singular de Investigación en Química Biolóxica
e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| | - Eduardo Fernandez-Megia
- Centro
Singular de Investigación en Química Biolóxica
e Materiais Moleculares (CIQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| |
Collapse
|
4
|
Chang YC, Shieh MC, Chang YH, Huang WL, Su WC, Cheng FY, Cheung CHA. Development of a cancer cells self‑activating and miR‑125a‑5p expressing poly‑pharmacological nanodrug for cancer treatment. Int J Mol Med 2022; 50:102. [PMID: 35703361 PMCID: PMC9239037 DOI: 10.3892/ijmm.2022.5158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer cells can acquire resistance to targeted therapeutic agents when the designated targets or their downstream signaling molecules develop protein conformational or activity changes. There is an increasing interest in developing poly-pharmacologic anticancer agents to target multiple oncoproteins or signaling pathways in cancer cells. The microRNA 125a-5p (miR-125a-5p) is a tumor suppressor, and its expression has frequently been downregulated in tumors. By contrast, the anti-apoptotic molecule BIRC5/SURVIVIN is highly expressed in tumors but not in the differentiated normal tissues. In the present study, the development of a BIRC5 gene promoter-driven, miR-125a-5p expressing, poly-L-lysine-conjugated magnetite iron poly-pharmacologic nanodrug (pL-MNP-pSur-125a) was reported. The cancer cells self-activating property and the anticancer effects of this nanodrug were examined in both the multidrug efflux protein ABCB1/MDR1-expressing/-non-expressing cancer cells in vitro and in vivo. It was demonstrated that pL-MNP-pSur-125a decreased the expression of ERBB2/HER2, HDAC5, BIRC5, and SP1, which are hot therapeutic targets for cancer in vitro. Notably, pL-MNP-pSur-125a also downregulated the expression of TDO2 in the human KB cervical carcinoma cells. PL-MNP-pSur-125a decreased the viability of various BIRC5-expressing cancer cells, regardless of the tissue origin or the expression of ABCB1, but not of the human BIRC5-non-expressing HMEC-1 endothelial cells. In vivo, pL-MNP-pSur-125a exhibited potent antitumor growth effects, but without inducing liver toxicity, in various zebrafish human-ABCB1-expressing and ABCB1-non-expressing tumor xenograft models. In conclusion, pL-MNP-pSur-125a is an easy-to-prepare and a promising poly-pharmacological anticancer nanodrug that has the potential to manage numerous malignancies, particularly for patients with BIRC5/ABCB1-related drug resistance after prolonged chemotherapeutic treatments.
Collapse
Affiliation(s)
- Yung-Chieh Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Min-Chieh Shieh
- Division of General Surgery, Department of Surgery, Ditmanson Medical Foundation Chia‑Yi Christian Hospital, Chiayi 600566, Taiwan, R.O.C
| | - Yen-Hsuan Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Wei-Lun Huang
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Wu-Chou Su
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Fong-Yu Cheng
- Department of Chemistry, College of Sciences, Chinese Culture University, Taipei 111396, Taiwan, R.O.C
| | - Chun Hei Antonio Cheung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| |
Collapse
|
5
|
Kang M, Lee SH, Kwon M, Byun J, Kim D, Kim C, Koo S, Kwon SP, Moon S, Jung M, Hong J, Go S, Song SY, Choi JH, Hyeon T, Oh YK, Park HH, Kim BS. Nanocomplex-Mediated In Vivo Programming to Chimeric Antigen Receptor-M1 Macrophages for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103258. [PMID: 34510559 DOI: 10.1002/adma.202103258] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/06/2021] [Indexed: 06/13/2023]
Abstract
Chimeric antigen receptor-T (CAR-T) cell immunotherapy has shown impressive clinical outcomes for hematologic malignancies. However, its broader applications are challenged due to its complex ex vivo cell-manufacturing procedures and low therapeutic efficacy against solid tumors. The limited therapeutic effects are partially due to limited CAR-T cell infiltration to solid tumors and inactivation of CAR-T cells by the immunosuppressive tumor microenvironment. Here, a facile approach is presented to in vivo program macrophages, which can intrinsically penetrate solid tumors, into CAR-M1 macrophages displaying enhanced cancer-directed phagocytosis and anti-tumor activity. In vivo injected nanocomplexes of macrophage-targeting nanocarriers and CAR-interferon-γ-encoding plasmid DNA induce CAR-M1 macrophages that are capable of CAR-mediated cancer phagocytosis, anti-tumor immunomodulation, and inhibition of solid tumor growth. Together, this study describes an off-the-shelf CAR-macrophage therapy that is effective for solid tumors and avoids the complex and costly processes of ex vivo CAR-cell manufacturing.
Collapse
Affiliation(s)
- Mikyung Kang
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seong Ho Lee
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Miji Kwon
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Junho Byun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sagang Koo
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seuk Young Song
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae Hyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul, 04763, Republic of Korea
- Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, Seoul, 04763, Republic of Korea
| | - Byung-Soo Kim
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
6
|
Lee YH, Kuo PW, Chen CJ, Sue CJ, Hsu YF, Pan MC. Indocyanine Green-Camptothecin Co-Loaded Perfluorocarbon Double-Layer Nanocomposite: A Versatile Nanotheranostics for Photochemotherapy and FDOT Diagnosis of Breast Cancer. Pharmaceutics 2021; 13:pharmaceutics13091499. [PMID: 34575572 PMCID: PMC8466706 DOI: 10.3390/pharmaceutics13091499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 01/10/2023] Open
Abstract
Breast cancer remains the most frequently diagnosed cancer and is the leading cause of neoplastic disease burden for females worldwide, suggesting that effective therapeutic and/or diagnostic strategies are still urgently needed. In this study, a type of indocyanine green (ICG) and camptothecin (CPT) co-loaded perfluorocarbon double-layer nanocomposite named ICPNC was developed for detection and photochemotherapy of breast cancer. The ICPNCs were designed to be surface modifiable for on-demand cell targeting and can serve as contrast agents for fluorescence diffuse optical tomography (FDOT). Upon near infrared (NIR) irradiation, the ICPNCs can generate a significantly increased production of singlet oxygen compared to free ICG, and offer a comparable cytotoxicity with reduced chemo-drug dosage. Based on the results of animal study, we further demonstrated that the ICPNCs ([ICG]/[CPT] = 40-/7.5-μM) in association with 1-min NIR irradiation (808 nm, 6 W/cm2) can provide an exceptional anticancer effect to the MDA-MB-231 tumor-bearing mice whereby the tumor size was significantly reduced by 80% with neither organ damage nor systemic toxicity after a 21-day treatment. Given a number of aforementioned merits, we anticipate that the developed ICPNC is a versatile theranostic nanoagent which is highly promising to be used in the clinic.
Collapse
Affiliation(s)
- Yu-Hsiang Lee
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City 320317, Taiwan; (P.-W.K.); (C.-J.C.); (C.-J.S.)
- Department of Chemical and Materials Engineering, National Central University, Taoyuan City 320317, Taiwan
- Correspondence: (Y.-H.L.); (M.-C.P.); Tel.: +886-3-422-7151 (ext. 27755) (Y.-H.L.); +886-3-422-7151 (ext. 34312) (M.-C.P.)
| | - Po-Wei Kuo
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City 320317, Taiwan; (P.-W.K.); (C.-J.C.); (C.-J.S.)
| | - Chun-Ju Chen
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City 320317, Taiwan; (P.-W.K.); (C.-J.C.); (C.-J.S.)
| | - Chu-Jih Sue
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City 320317, Taiwan; (P.-W.K.); (C.-J.C.); (C.-J.S.)
| | - Ya-Fen Hsu
- Department of Surgery, Landseed International Hospital, Taoyuan City 324609, Taiwan;
| | - Min-Chun Pan
- Department of Mechanical Engineering, National Central University, Taoyuan City 320317, Taiwan
- Correspondence: (Y.-H.L.); (M.-C.P.); Tel.: +886-3-422-7151 (ext. 27755) (Y.-H.L.); +886-3-422-7151 (ext. 34312) (M.-C.P.)
| |
Collapse
|
7
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
8
|
Recent advances in peptide-targeted micelleplexes: Current developments and future perspectives. Int J Pharm 2021; 597:120362. [PMID: 33556489 DOI: 10.1016/j.ijpharm.2021.120362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
The decoding of the human genome revolutionized the understanding of how genetics influence the interplay between health and disease, in a multidisciplinary perspective. Thus, the development of exogenous nucleic acids-based therapies has increased to overcome hereditary or acquired genetic-associated diseases. Gene drug delivery using non-viral systems, for instance micelleplexes, have been recognized as promising options for gene-target therapies. Micelleplexes are core-shell structures, at a nanometric scale, designed using amphiphilic block copolymers. These can self-assemble in an aqueous medium, leading to the formation of a hydrophilic and positively charged corona - that can transport nucleic acids, - and a hydrophobic core - which can transport poor water-soluble drugs. However, the performance of these types of carriers usually is hindered by several in vivo barriers. Fortunately, due to a significant amount of research, strategies to overcome these shortcomings emerged. With a wide range of structural features, good stability against proteolytic degradation, affordable characteristic, easy synthesis, low immunogenicity, among other advantages, peptides have increasingly gained popularity as target ligands for non-viral carriers. Hence, this review addresses the use of peptides with micelleplexes illustrating, through the analysis of in vitro and in vivo studies, the potential and future perspectives of this combination.
Collapse
|
9
|
Richter F, Mapfumo P, Martin L, Solomun JI, Hausig F, Frietsch JJ, Ernst T, Hoeppener S, Brendel JC, Traeger A. Improved gene delivery to K-562 leukemia cells by lipoic acid modified block copolymer micelles. J Nanobiotechnology 2021; 19:70. [PMID: 33676500 PMCID: PMC7936509 DOI: 10.1186/s12951-021-00801-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/09/2021] [Indexed: 12/23/2022] Open
Abstract
Although there has been substantial progress in the research field of gene delivery, there are some challenges remaining, e.g. there are still cell types such as primary cells and suspension cells (immune cells) known to be difficult to transfect. Cationic polymers have gained increasing attention due to their ability to bind, condense and mask genetic material, being amenable to scale up and highly variable in their composition. In addition, they can be combined with further monomers exhibiting desired biological and chemical properties, such as antioxidative, pH- and redox-responsive or biocompatible features. By introduction of hydrophobic monomers, in particular as block copolymers, cationic micelles can be formed possessing an improved chance of transfection in otherwise challenging cells. In this study, the antioxidant biomolecule lipoic acid, which can also be used as crosslinker, was incorporated into the hydrophobic block of a diblock copolymer, poly{[2-(dimethylamino)ethyl methacrylate]101-b-[n-(butyl methacrylate)124-co-(lipoic acid methacrylate)22]} (P(DMAEMA101-b-[nBMA124-co-LAMA22])), synthesized by RAFT polymerization and assembled into micelles (LAMA-mic). These micelles were investigated regarding their pDNA binding, cytotoxicity mechanisms and transfection efficiency in K-562 and HEK293T cells, the former representing a difficult to transfect, suspension leukemia cell line. The LAMA-mic exhibited low cytotoxicity at applied concentrations but demonstrated superior transfection efficiency in HEK293T and especially K-562 cells. In-depth studies on the transfection mechanism revealed that transfection efficiency in K-562 cells does not depend on the specific oncogenic fusion gene BCR-ABL alone. It is independent of the cellular uptake of polymer-pDNA complexes but correlates with the endosomal escape of the LAMA-mic. A comparison of the transfection efficiency of the LAMA-mic with structurally comparable micelles without lipoic acid showed that lipoic acid is not solely responsible for the superior transfection efficiency of the LAMA-mic. More likely, a synergistic effect of the antioxidative lipoic acid and the micellar architecture was identified. Therefore, the incorporation of lipoic acid into the core of hydrophobic-cationic micelles represents a promising tailor-made transfer strategy, which can potentially be beneficial for other difficult to transfect cell types.
Collapse
Affiliation(s)
- Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Prosper Mapfumo
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Liam Martin
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jana I Solomun
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Franziska Hausig
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jochen J Frietsch
- Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Thomas Ernst
- Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany.
| |
Collapse
|
10
|
Wang X, Qiu Y, Wang M, Zhang C, Zhang T, Zhou H, Zhao W, Zhao W, Xia G, Shao R. Endocytosis and Organelle Targeting of Nanomedicines in Cancer Therapy. Int J Nanomedicine 2020; 15:9447-9467. [PMID: 33268987 PMCID: PMC7701161 DOI: 10.2147/ijn.s274289] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Nanomedicines (NMs) have played an increasing role in cancer therapy as carriers to efficiently deliver therapeutics into tumor cells. For this application, the uptake of NMs by tumor cells is usually a prerequisite to deliver the cargo to intracellular locations, which mainly relies on endocytosis. NMs can enter cells through a variety of endocytosis pathways. Different endocytosis pathways exhibit different intracellular trafficking routes and diverse subcellular localizations. Therefore, a comprehensive understanding of endocytosis mechanisms is necessary for increasing cellular entry efficiency and to trace the fate of NMs after internalization. This review focuses on endocytosis pathways of NMs in tumor cells, mainly including clathrin- and caveolae-mediated endocytosis pathways, involving effector molecules, expression difference of those molecules between normal and tumor cells, as well as the intracellular trafficking route of corresponding endocytosis vesicles. Then, the latest strategies for NMs to actively employ endocytosis are described, including improving tumor cellular uptake of NMs by receptor-mediated endocytosis, transporter-mediated endocytosis and enabling drug activity by changing intracellular routes. Finally, active targeting strategies towards intracellular organelles are also mentioned. This review will be helpful not only in explicating endocytosis and the trafficking process of NMs and elucidating anti-tumor mechanisms inside the cell but also in rendering new ideas for the design of highly efficacious and cancer-targeted NMs.
Collapse
Affiliation(s)
- Xiaowei Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Yuhan Qiu
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Mengyan Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Conghui Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Tianshu Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Huimin Zhou
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wenxia Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wuli Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Rongguang Shao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
11
|
Zhang SK, Gong L, Zhang X, Yun ZM, Li SB, Gao HW, Dai CJ, Yuan JJ, Chen JM, Gong F, Tan YX, Ji SP. Antimicrobial peptide AR-23 derivatives with high endosomal disrupting ability enhance poly(l-lysine)-mediated gene transfer. J Gene Med 2020; 22:e3259. [PMID: 32776410 PMCID: PMC7685122 DOI: 10.1002/jgm.3259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/06/2020] [Accepted: 08/06/2020] [Indexed: 01/24/2023] Open
Abstract
Background pH‐sensitive peptides are a relatively new strategy for conquering the poor endosomal release of cationic polymer‐mediated transfection. Modification of antimicrobial peptides by exchanging positively‐charged residues with negatively‐charged glutamic acid residues (Glu) greatly improves its lytic activity at the endosomal pH, which could improve cationic polymer‐mediated transfection. Methods In the present study, we investigated the effect of the number of Glu substituted for positively‐charged residues on the endosomal escape activity of AR‐23 and the ability of mutated AR‐23 with respect to enhancing cationic polymer‐mediated transfection. Three analogs were synthesized by replacing the positively‐charged residues in the AR‐23 sequence with Glu one‐by‐one. Results The pH‐sensitive lysis ability of the peptides, the effect of peptides on the physicochemical characteristics, the intracellular trafficking, the transfection efficiency and the cytotoxicity of the polyplexes were determined. Increased lytic activity of peptides was observed with the increased number of Glu replacement in the AR‐23 sequence at acidic pH. The number of Glu substituted for positively‐charged residues of AR‐23 dramatically affects its lysis ability at neutral pH. Triple‐Glu substitution in the AR‐23 sequence greatly improved poly(l‐lysine)‐mediated gene transfection efficiency at the same time as maintaining low cytotoxicity. Conclusions The results indicate that replacement of positively‐charged residues with sufficient Glu residues may be considered as a method for designing pH‐sensitive peptides, which could be applied as potential enhancers for improving cationic polymer‐mediated transfection.
Collapse
Affiliation(s)
- Shi-Kun Zhang
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Lin Gong
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China.,PLA navy No. 971 Hospital, Qingdao, Shandong, China
| | - Xue Zhang
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Zhi-Min Yun
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Su-Bo Li
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Hong-Wei Gao
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Cong-Jie Dai
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian, China
| | - Jian-Jun Yuan
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian, China
| | - Jing-Ming Chen
- Quanzhou Preschool Education College, Quanzhou, Fujian, China
| | - Feng Gong
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Ying-Xia Tan
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Shou-Ping Ji
- Department of Stem Cell and Regenerative Medicine, Institute of Health Service and Transfusion Medicine, Beijing, China.,College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou, Fujian, China
| |
Collapse
|
12
|
Pal Singh P, Vithalapuram V, Metre S, Kodipyaka R. Lipoplex-based therapeutics for effective oligonucleotide delivery: a compendious review. J Liposome Res 2019; 30:313-335. [DOI: 10.1080/08982104.2019.1652645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Pirthi Pal Singh
- Department of Formulation Research and Development, Custom Pharmaceutical Services, Dr. Reddy’s Laboratories Ltd., Hyderabad, India
| | - Veena Vithalapuram
- Department of Formulation Research and Development, Custom Pharmaceutical Services, Dr. Reddy’s Laboratories Ltd., Hyderabad, India
| | - Sunita Metre
- Department of Formulation Research and Development, Custom Pharmaceutical Services, Dr. Reddy’s Laboratories Ltd., Hyderabad, India
| | - Ravinder Kodipyaka
- Department of Formulation Research and Development, Custom Pharmaceutical Services, Dr. Reddy’s Laboratories Ltd., Hyderabad, India
| |
Collapse
|
13
|
Zhang Q, Lu Y, Xu X, Li S, Du Y, Yu R. MR molecular imaging of HCC employing a regulated ferritin gene carried by a modified polycation vector. Int J Nanomedicine 2019; 14:3189-3201. [PMID: 31118631 PMCID: PMC6504634 DOI: 10.2147/ijn.s191270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/19/2019] [Indexed: 01/03/2023] Open
Abstract
Purpose: Early diagnosis is essential for reducing liver cancer mortality, and molecular diagnosis by magnetic resonance imaging (MRI) is an emerging and promising technology. The chief aim of the present work is to use the ferritin gene, modified by the alpha-fetoprotein (AFP) promoter, carried by a highly safe vector, to produce signal contrast on T2-weighted MR imaging as an endogenous contrast agent, and to provide a highly specific target for subsequent therapy. Methods: Polyethyleneimine-β-cyclodextrin (PEI-β-CD, PC) was synthesized as a novel vector. The optimal nitrogen/phosphorus ratio (N/P) of the PC/plasmid DNA complex was determined by gel retardation, biophysical properties and transmission electron microscopy morphological analysis. The transfection efficiency was observed under a fluorescence microscope and analyzed by flow cytometry. Cellular iron accumulation caused by ferritin overexpression was verified by Prussian blue staining, and the resulting contrast imaging effect was examined by MRI. Results: The modified cationic polymer PC was much safer than high molecular weight PEI, and could condense plasmid DNA at an N/P ratio of 50 with suitable biophysical properties and a high transfection efficiency. Overexpression of ferritin enriched intracellular iron. The short-term iron imbalance initiated by AFP promoter regulation only occurred in hepatoma cells, resulting in signal contrast on MRI. The specific target TfR was also upregulated during this process. Conclusion: These results illustrate that the regulated ferritin gene carried by PC can be used as an endogenous contrast agent for MRI detection of hepatocellular carcinoma (HCC). This molecular imaging technique may promote safer early diagnosis of HCC, and provide a more highly specific target for future chemotherapy drugs.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yuanfei Lu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaoling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Shujuan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Risheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
14
|
Wojnilowicz M, Glab A, Bertucci A, Caruso F, Cavalieri F. Super-resolution Imaging of Proton Sponge-Triggered Rupture of Endosomes and Cytosolic Release of Small Interfering RNA. ACS NANO 2019; 13:187-202. [PMID: 30566836 DOI: 10.1021/acsnano.8b05151] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The intracellular delivery of nucleic acids and proteins remains a key challenge in the development of biological therapeutics. In gene therapy, the inefficient delivery of small interfering RNA (siRNA) to the cytosol by lipoplexes or polyplexes is often ascribed to the entrapment and degradation of siRNA payload in the endosomal compartments. A possible mechanism by which polyplexes rupture the endosomal membrane and release their nucleic acid cargo is commonly defined as the "proton sponge effect". This is an osmosis-driven process triggered by the proton buffering capacity of polyplexes. Herein, we investigate the molecular basis of the "proton sponge effect" through direct visualization of the siRNA trafficking process, including analysis of individual polyplexes and endosomes, using stochastic optical reconstruction microscopy. We probe the sequential siRNA trafficking steps through single molecule super-resolution analysis of subcellular structures, polyplexes, and silencing RNA molecules. Specifically, individual intact polyplexes released in the cytosol upon rupture of the endosomes, the damaged endosomal vesicles, and the disassembly of the polyplexes in the cytosol are examined. We find that the architecture of the polyplex and the rigidity of the cationic polymer chains are crucial parameters that control the mechanism of endosomal escape driven by the proton sponge effect. We provide evidence that in highly branched and rigid cationic polymers, such as glycogen or polyethylenimine, immobilized on silica nanoparticles, the proton sponge effect is effective in inducing osmotic swelling and rupture of endosomes.
Collapse
Affiliation(s)
- Marcin Wojnilowicz
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering , The University of Melbourne , Parkville , Victoria 3010 , Australia
| | - Agata Glab
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering , The University of Melbourne , Parkville , Victoria 3010 , Australia
| | - Alessandro Bertucci
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering , The University of Melbourne , Parkville , Victoria 3010 , Australia
- Dipartimento di Scienze e Tecnologie Chimiche , Universita' degli Studi di Roma "Tor Vergata" , via della Ricerca Scientifica 1 , 00133 Rome , Italy
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering , The University of Melbourne , Parkville , Victoria 3010 , Australia
| | - Francesca Cavalieri
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering , The University of Melbourne , Parkville , Victoria 3010 , Australia
- Dipartimento di Scienze e Tecnologie Chimiche , Universita' degli Studi di Roma "Tor Vergata" , via della Ricerca Scientifica 1 , 00133 Rome , Italy
| |
Collapse
|
15
|
Shende P, Ture N, Gaud RS, Trotta F. Lipid- and polymer-based plexes as therapeutic carriers for bioactive molecules. Int J Pharm 2019; 558:250-260. [PMID: 30641179 DOI: 10.1016/j.ijpharm.2018.12.085] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/25/2018] [Accepted: 12/27/2018] [Indexed: 12/16/2022]
Abstract
Recently, promising strategies of plexes include the complexation of nucleic acids with lipids (lipoplexes) and different kinds of polymers (polyplexes) for delivery of actives and genetic material in abnormal conditions like cancer, cystic fibrosis and genetic disorders. The present review article focuses on the comparative aspects of lipoplexes and polyplexes associated with molecular structure, cellular transportation and formulation aspects. The major advantages of lipoplexes and polyplexes over conventional liposomes involve non-immunogenic viral gene transfer, facile manufacturing and preservation of genetic material encapsulated within the nanocarriers. Lipoplexes and polyplexes enhance the transfection of DNA into the cell by stepwise electrostatic cationic-anionic interaction with DNA backbones. The ease and cost-effective formation of complexes extend their applications in the treatment of cancer and genetic disorders. Lipoplexes and polyplexes necessitate intensive research in the fields of quality, toxicity and methods of preparation for commercialization.
Collapse
Affiliation(s)
- Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L Mehta Road, Vile Parle (W), Mumbai, India.
| | - Narayan Ture
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L Mehta Road, Vile Parle (W), Mumbai, India
| | - R S Gaud
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L Mehta Road, Vile Parle (W), Mumbai, India
| | - F Trotta
- Department of Chemistry, University of Torino, Italy
| |
Collapse
|
16
|
Rödl W, Taschauer A, Schaffert D, Wagner E, Ogris M. Synthesis of Polyethylenimine-Based Nanocarriers for Systemic Tumor Targeting of Nucleic Acids. Methods Mol Biol 2019; 1943:83-99. [PMID: 30838611 DOI: 10.1007/978-1-4939-9092-4_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Nucleic acid-based therapies offer the option to treat tumors in a highly selective way, while toxicity towards healthy tissue can be avoided when proper delivery vehicles are used. We have recently developed carrier systems based on linear polyethylenimine, which after chemical coupling of protein- or peptide-based ligands can form nanosized polyplexes with plasmid DNA (pDNA) or RNA and deliver their payload into target cells by receptor-mediated endocytosis. This chapter describes the synthesis of LPEI from a precursor polymer and the current coupling techniques and purification procedure for peptide conjugates with linear polyethylenimine. A protocol is also given for the formation and characterization of polyplexes formed with LPEI conjugate and pDNA.
Collapse
Affiliation(s)
- Wolfgang Rödl
- Pharmaceutical Biotechnology, Center for System Based Drug Research, Ludwig-Maximilians-University, Munich, Germany
| | - Alexander Taschauer
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Department of Pharmaceutical Chemistry, Center of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - David Schaffert
- Department of Molecular Biology, Aarhus University, Aarhus, Denmark
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System Based Drug Research, Ludwig-Maximilians-University, Munich, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-University, Munich, Germany
| | - Manfred Ogris
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Department of Pharmaceutical Chemistry, Center of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria.
- Center for NanoScience (CeNS), Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
17
|
Ahn J, Cho CS, Cho SW, Kang JH, Kim SY, Min DH, Song JM, Park TE, Jeon NL. Investigation on vascular cytotoxicity and extravascular transport of cationic polymer nanoparticles using perfusable 3D microvessel model. Acta Biomater 2018; 76:154-163. [PMID: 29807185 DOI: 10.1016/j.actbio.2018.05.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/16/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022]
Abstract
Vascular networks are the first sites exposed to cationic polymer nanoparticles (NPs) administered intravenously, and thus function as a barrier for NPs reaching the target organ. While cationic polymer NPs have been intensively studied as non-viral delivery systems, their biological effects in human microvessels have been poorly investigated due to a lack of appropriate in vitro systems. Here, we employed a three-dimensional microvessel on a chip, which accurately models in vivo conditions. An open and perfused microvessel surrounded by pericytes was shown to reproduce the important features of living vasculature, including barrier function and biomarkers. Using this microvessel chip, we observed contraction of the microvascular lumen induced by perfused polyethylenimine (PEI)/DNA NPs. We demonstrated that the oxidative stress present when microvessels were exposed to PEI NPs led to rearrangement of microtubules resulting in microvessel contraction. Furthermore, the transcytotic behavior of PEI NPs was analyzed in the microvessel by monitoring the escape of PEI NPs from the microvascular lumen into the perivascular region, which was not possible in two-dimensional culture systems. With our new understanding of the different behaviors of cationic polymer NPs depending on their transcytotic route, we suggest that caveolae-mediated transcytosis is a powerful route for efficient extravascular transport. STATEMENT OF SIGNIFICANCE Microvascular networks are not only biological system constituting largest surface area in the body and but also first site exposed to nanoparticle in vivo. While cationic polymer NPs have been intensively studied as non-viral delivery systems, its biological effects in human microvessel have been poorly investigated due to lack of appropriate in vitro systems. Here, we microengineered an open and perfused 3D pericyte incorporated microvessel model which possesses same morphological characteristic of in vivo. Using the microengineered model, this study represents the first report of transcytotic behavior of NPs in 3D microvessel, and its effect on extravasation efficiency. Our study lays the groundwork for the integration of innovative technologies to examine blood vessel-nanoparticle interaction, which a critical but ill-defined phenomenon.
Collapse
Affiliation(s)
- Jungho Ahn
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 08826, South Korea; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA 30332, USA
| | - Chong-Su Cho
- Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Seong Woo Cho
- Ulsan National Institute of Science and Technology, Ulsan 44914, South Korea
| | - Joo H Kang
- Ulsan National Institute of Science and Technology, Ulsan 44914, South Korea
| | - Sung-Yon Kim
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, South Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Joon Myong Song
- College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Tae-Eun Park
- Ulsan National Institute of Science and Technology, Ulsan 44914, South Korea.
| | - Noo Li Jeon
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
18
|
Zhang H, De Smedt SC, Remaut K. Fluorescence Correlation Spectroscopy to find the critical balance between extracellular association and intracellular dissociation of mRNA complexes. Acta Biomater 2018; 75:358-370. [PMID: 29753914 DOI: 10.1016/j.actbio.2018.05.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/13/2018] [Accepted: 05/09/2018] [Indexed: 12/20/2022]
Abstract
Fluorescence Correlation Spectroscopy (FCS) is a promising tool to study interactions on a single molecule level. The diffusion of fluorescent molecules in and out of the excitation volume of a confocal microscope leads to the fluorescence fluctuations that give information on the average number of fluorescent molecules present in the excitation volume and their diffusion coefficients. In this context, we complexed mRNA into lipoplexes and polyplexes and explored the association/dissociation degree of complexes by using gel electrophoresis and FCS. FCS enabled us to measure the association and dissociation degree of mRNA-based complexes both in buffer and protein-rich biological fluids such as human serum and ascitic fluid, which is a clear advantage over gel electrophoresis that was only applicable in protein-free buffer solutions. Furthermore, following the complex stability in buffer and biological fluids by FCS assisted to understand how complex characteristics, such as charge ratio and strength of mRNA binding, correlated to the transfection efficiency. We found that linear polyethyleneimine prevented efficient translation of mRNA, most likely due to a too strong mRNA binding, whereas the lipid based carrier Lipofectamine® messengerMAX did succeed in efficient release and subsequent translation of mRNA in the cytoplasm of the cells. Overall, FCS is a reliable tool for the in depth characterization of mRNA complexes and can help us to find the critical balance keeping mRNA bound in complexes in the extracellular environment and efficient intracellular mRNA release leading to protein production. STATEMENT OF SIGNIFICANCE The delivery of messenger RNA (mRNA) to cells is promising to treat a variety of diseases. Therefore, the mRNA is typically packed in small lipid particles or polymer particles that help the mRNA to reach the cytoplasm of the cells. These particles should bind and carry the mRNA in the extracellular environment (e.g. blood, peritoneal fluid, …), but should release the mRNA again in the intracellular environment. In this paper, we evaluated a method (Fluorescence Correlation Spectroscopy) that allows for the in depth characterization of mRNA complexes and can help us to find the critical balance keeping mRNA bound in complexes in the extracellular environment and efficient intracellular mRNA release leading to protein production.
Collapse
|
19
|
Zhang Z, Wan T, Chen Y, Chen Y, Sun H, Cao T, Songyang Z, Tang G, Wu C, Ping Y, Xu FJ, Huang J. Cationic Polymer-Mediated CRISPR/Cas9 Plasmid Delivery for Genome Editing. Macromol Rapid Commun 2018; 40:e1800068. [PMID: 29708298 DOI: 10.1002/marc.201800068] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/19/2018] [Indexed: 12/12/2022]
Abstract
Delivery of CRISPR (clustered regularly interspaced short palindromic repeats)/CRISPR-associated protein-9 (Cas9) represents a major hurdle for successful clinical translation of genome editing tools. Owing to the large size of plasmids that encode Cas9 and single-guide RNA (sgRNA), genome editing efficiency mediated by current delivery carriers is still unsatisfactory to meet the requirement for its real applications. Herein, cationic polymer polyethyleneimine-β-cyclodextrin (PC), known to be efficient for small plasmid transfection, is reported to likewise mediate efficient delivery of plasmid encoding Cas9 and sgRNA. Whereas PC can condense and encapsulate large plasmids at high N/P ratio, the delivery of plasmid results in efficient editing at two genome loci, namely, hemoglobin subunit beta (19.1%) and rhomboid 5 homolog 1 (RHBDF1) (7.0%). Sanger sequencing further confirms the successful genome editing at these loci. This study defines a new strategy for the delivery of the large plasmid encoding Cas9/sgRNA for efficient genome editing.
Collapse
Affiliation(s)
- Zhen Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, Guangzhou Key Laboratory of Healthy Aging Research and State Key Laboratory of Biocontrol, SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Tao Wan
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Institute of Pharmaceutics, College of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yuxuan Chen
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Yu Chen
- Key Laboratory of Gene Engineering of the Ministry of Education, Guangzhou Key Laboratory of Healthy Aging Research and State Key Laboratory of Biocontrol, SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Hongwei Sun
- Key Laboratory of Gene Engineering of the Ministry of Education, Guangzhou Key Laboratory of Healthy Aging Research and State Key Laboratory of Biocontrol, SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Tianqi Cao
- Key Laboratory of Gene Engineering of the Ministry of Education, Guangzhou Key Laboratory of Healthy Aging Research and State Key Laboratory of Biocontrol, SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Zhou Songyang
- Key Laboratory of Gene Engineering of the Ministry of Education, Guangzhou Key Laboratory of Healthy Aging Research and State Key Laboratory of Biocontrol, SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510275, China.,Key Laboratory of Reproductive Medicine of Guangdong Province, School of Life Sciences and the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| | - Guping Tang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310028, China
| | - Chuanbin Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yuan Ping
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Fu-Jian Xu
- Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Junjiu Huang
- Key Laboratory of Gene Engineering of the Ministry of Education, Guangzhou Key Laboratory of Healthy Aging Research and State Key Laboratory of Biocontrol, SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510275, China.,Key Laboratory of Reproductive Medicine of Guangdong Province, School of Life Sciences and the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| |
Collapse
|
20
|
Lee YH, Lin YC. Anti-EGFR Indocyanine Green-Mitomycin C-Loaded Perfluorocarbon Double Nanoemulsion: A Novel Nanostructure for Targeted Photochemotherapy of Bladder Cancer Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E283. [PMID: 29701711 PMCID: PMC5977297 DOI: 10.3390/nano8050283] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/09/2018] [Accepted: 04/25/2018] [Indexed: 01/14/2023]
Abstract
The use of phototherapy as an adjuvant bladder cancer treatment has long been considered, but its application has been severely hampered due to a lack of tumor specificity, unpredicted cytotoxicity, and insufficient anticancer efficacy. In this study, we aim to manufacture anti-EGFR indocyanine green (ICG) mitomycin C (MMC) encapsulated perfluorocarbon double nanoemulsions (EIMPDNEs), and explore their photochemotherapeutic efficacy on EGFR-expressing bladder cancer cells in vitro. The EIMPDNEs were manufactured using a double emulsification technique followed by antibody conjugation on the particles’ surfaces. The EIMPDNE were 257 ± 19.4 nm in size, with a surface charge of −12.3 ± 2.33 mV. The EGFR targetability of the EIMPNDE was confirmed by its enhanced binding efficiency to T24 cells when compared with the performance of nanodroplets without EGFR conjugation (p < 0.05). In comparison with freely dissolved ICG, the EIMPDNEs with equal ICG content conferred an improved thermal stability to the encapsulated ICG, and were able to provide a comparable hyperthermia effect and significantly enhanced the production of singlet oxygen under 808 nm near infrared (NIR) exposure with an intensity of 6 W cm−2 for 5 min (p < 0.05). Based on viability analyses, our data showed that the EIMPDNEs were effective in bladder cancer cell eradication upon NIR exposure (808 nm; 6 W cm−2), and the resulting cell death rate was even higher than that caused by a five-fold higher amount of entrapped MMC alone. With the merits of improved ICG stability, EGFR binding specificity, and effective cancer cell eradication, the EIMPDNEs exhibit potential for use in EGFR-expressing bladder cancer therapy with lower chemotoxicity.
Collapse
Affiliation(s)
- Yu-Hsiang Lee
- Department of Biomedical Sciences and Engineering, National Central University. No. 300, Jhongda Rd., Taoyuan City 32001, Taiwan.
- Department of Chemical and Materials Engineering, National Central University, Taoyuan City 32001, Taiwan.
| | - Yu-Chun Lin
- Department of Biomedical Sciences and Engineering, National Central University. No. 300, Jhongda Rd., Taoyuan City 32001, Taiwan.
| |
Collapse
|
21
|
Vermeulen LMP, Brans T, Samal SK, Dubruel P, Demeester J, De Smedt SC, Remaut K, Braeckmans K. Endosomal Size and Membrane Leakiness Influence Proton Sponge-Based Rupture of Endosomal Vesicles. ACS NANO 2018; 12:2332-2345. [PMID: 29505236 DOI: 10.1021/acsnano.7b07583] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
In gene therapy, endosomal escape represents a major bottleneck since nanoparticles often remain entrapped inside endosomes and are trafficked toward the lysosomes for degradation. A detailed understanding of the endosomal barrier would be beneficial for developing rational strategies to improve transfection and endosomal escape. By visualizing individual endosomal escape events in live cells, we obtain insight into mechanistic factors that influence proton sponge-based endosomal escape. In a comparative study, we found that HeLa cells treated with JetPEI/pDNA polyplexes have a 3.5-fold increased endosomal escape frequency compared to ARPE-19 cells. We found that endosomal size has a major impact on the escape capacity. The smaller HeLa endosomes are more easily ruptured by the proton sponge effect than the larger ARPE-19 endosomes, a finding supported by a mathematical model based on the underlying physical principles. Still, it remains intriguing that even in the small HeLa endosomes, <10% of the polyplex-containing endosomes show endosomal escape. Further experiments revealed that the membrane of polyplex-containing endosomes becomes leaky to small compounds, preventing effective buildup of osmotic pressure, which in turn prevents endosomal rupture. Analysis of H1299 and A549 cells revealed that endosomal size determines endosomal escape efficiency when cells have comparable membrane leakiness. However, at high levels of membrane leakiness, buildup of osmotic pressure is no longer possible, regardless of endosomal size. Based on our findings that both endosomal size and membrane leakiness have a high impact on proton sponge-based endosomal rupture, we provide important clues toward further improvement of this escape strategy.
Collapse
Affiliation(s)
- Lotte M P Vermeulen
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Toon Brans
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Sangram K Samal
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | | | - Jo Demeester
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Katrien Remaut
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy and ‡Centre for Nano- and Biophotonics , Ghent University , Ottergemsesteenweg 460 , 9000 Ghent , Belgium
| |
Collapse
|
22
|
Wong LY, Xia B, Wolvetang E, Cooper-White J. Targeted, Stimuli-Responsive Delivery of Plasmid DNA and miRNAs Using a Facile Self-Assembled Supramolecular Nanoparticle System. Biomacromolecules 2018; 19:353-363. [DOI: 10.1021/acs.biomac.7b01462] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
| | | | | | - Justin Cooper-White
- Biomedical
Manufacturing, CSIRO, Monash University, Clayton, VIC 3069, Australia
- School
of Chemical Engineering, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
23
|
Mastorakos P, Zhang C, Song E, Kim YE, Park HW, Berry S, Choi WK, Hanes J, Suk JS. Biodegradable brain-penetrating DNA nanocomplexes and their use to treat malignant brain tumors. J Control Release 2017; 262:37-46. [PMID: 28694032 DOI: 10.1016/j.jconrel.2017.07.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/21/2017] [Accepted: 07/07/2017] [Indexed: 11/30/2022]
Abstract
The discovery of powerful genetic targets has spurred clinical development of gene therapy approaches to treat patients with malignant brain tumors. However, lack of success in the clinic has been attributed to the inability of conventional gene vectors to achieve gene transfer throughout highly disseminated primary brain tumors. Here, we demonstrate ex vivo that small nanocomplexes composed of DNA condensed by a blend of biodegradable polymer, poly(β-amino ester) (PBAE), with PBAE conjugated with 5kDa polyethylene glycol (PEG) molecules (PBAE-PEG) rapidly penetrate healthy brain parenchyma and orthotopic brain tumor tissues in rats. Rapid diffusion of these DNA-loaded nanocomplexes observed in fresh tissues ex vivo demonstrated that they avoided adhesive trapping in the brain owing to their dense PEG coating, which was critical to achieving widespread transgene expression throughout orthotopic rat brain tumors in vivo following administration by convection enhanced delivery. Transgene expression with the PBAE/PBAE-PEG blended nanocomplexes (DNA-loaded brain-penetrating nanocomplexes, or DNA-BPN) was uniform throughout the tumor core compared to nanocomplexes composed of DNA with PBAE only (DNA-loaded conventional nanocomplexes, or DNA-CN), and transgene expression reached beyond the tumor edge, where infiltrative cancer cells are found, only for the DNA-BPN formulation. Finally, DNA-BPN loaded with anti-cancer plasmid DNA provided significantly enhanced survival compared to the same plasmid DNA loaded in DNA-CN in two aggressive orthotopic brain tumor models in rats. These findings underscore the importance of achieving widespread delivery of therapeutic nucleic acids within brain tumors and provide a promising new delivery platform for localized gene therapy in the brain.
Collapse
Affiliation(s)
- Panagiotis Mastorakos
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clark Zhang
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric Song
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Young Eun Kim
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Hee Won Park
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sneha Berry
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Won Kyu Choi
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Justin Hanes
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Oncology, Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Jung Soo Suk
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
24
|
Krzysztoń R, Salem B, Lee DJ, Schwake G, Wagner E, Rädler JO. Microfluidic self-assembly of folate-targeted monomolecular siRNA-lipid nanoparticles. NANOSCALE 2017; 9:7442-7453. [PMID: 28530287 DOI: 10.1039/c7nr01593c] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Non-viral delivery of nucleic acids for therapies based on RNA interference requires a rational design and optimal self-assembly strategies. Nucleic acid particles need to be small, stable and functional in terms of selective cell uptake and controlled release of encapsulated nucleic acids. Here we report on small (∼38 nm) monomolecular nucleic acid/lipid particles (mNALPs) that contain single molecules of short double-stranded oligonucleotides covered by a tight, highly curved lipid bilayer. The particles consist of DOPE, DOTAP, DOPC and DSPE-PEG(2000) and are assembled with 21 bp double-stranded DNA or small interfering RNA by solvent exchange on a hydrodynamic-focusing microfluidic chip. In comparison to vortex mixing by hand this method increases the encapsulation efficiency by 20%, and yields particles with a narrower size distribution, negligible aggregate formation and high stability in blood plasma and serum. Modification of mNALPs with folate-conjugated PEG-lipids results in specific binding and uptake by epithelial carcinoma KB cells overexpressing folate receptors. Binding is significantly reduced by competitive inhibition using free folate and is not observed with non-targeted mNALPs, revealing high specificity. The functionalized mNALPs show gene silencing in the presence of chloroquine, an endosome-destabilizing agent. Together, the robust self-assembly of small-sized mNALPs with their high stability and receptor-specific cell uptake demonstrate that the tight, PEG-grafted lipid-bilayer encapsulation may offer a promising approach towards the delivery of short double-stranded oligonucleotides.
Collapse
Affiliation(s)
- R Krzysztoń
- Faculty of Physics, Ludwig-Maximilians-Universität Munich (LMU), Geschwister-Scholl-Platz 1, Munich 80539, Germany
| | | | | | | | | | | |
Collapse
|
25
|
Lee YH, Ma YT. Synthesis, characterization, and biological verification of anti-HER2 indocyanine green-doxorubicin-loaded polyethyleneimine-coated perfluorocarbon double nanoemulsions for targeted photochemotherapy of breast cancer cells. J Nanobiotechnology 2017; 15:41. [PMID: 28521752 PMCID: PMC5437512 DOI: 10.1186/s12951-017-0274-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/08/2017] [Indexed: 11/22/2022] Open
Abstract
Background Breast cancer is the most frequently diagnosed cancer and the leading cause of cancer death among females worldwide. Among various types of breast cancer, the human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer is known to be more aggressive and often resistant to medicinal treatment, leading to an insufficient prognosis and poor susceptibility to chemotherapy and/or hormonal therapy in the current clinic. These circumstances implicate that developing an improved therapeutic strategy rather than persistently changing the anticancer drugs for trying is truly needed to successfully cure this type of breast cancer. In this study, we aimed to fabricate anti-HER2 indocyanine green (ICG)–doxorubicin (DOX)-loaded polyethyleneimine-coated perfluorocarbon double nanoemulsions (HIDPPDNEs) to explore the co-administration of phototherapy and chemotherapy for HER2-overexpressing breast cancer in vitro. Results The HIDPPDNE was first characterized as a sphere-like nanoparticle with surface charge of −57.1 ± 5.6 mV and size of 340.6 ± 4.5 nm, whereas the DOX release rates for the nanodroplets within 48 h in 4 and 37 °C were obtained by 8.13 ± 2.46% and 19.88 ± 2.75%, respectively. We then examined the target-ability of the nanostructure and found that the adhesion efficiency of the HIDPPDNEs onto HER2+ MDA-MB-453 cells was threefold higher than the nanodroplets without anti-HER2 antibody, indicating that the HIDPPDNEs are the product with HER2 binding specificity. In comparison to freely dissolved ICG, the HIDPPDNEs conferred an enhanced thermal stability to the entrapped ICG, and were able to provide a comparable hyperthermia effect and markedly increased production of singlet oxygen under near infrared irradiation (808 nm; 6 W/cm2). Based on the viability analyses, the results showed that the HIDPPDNEs were effective on cell eradication upon near infrared irradiation (808 nm; 6 W/cm2), and the resulting cell mortality was even higher than that caused by using twice amount of encapsulated DOX or ICG alone. Conclusions This work demonstrates that the HIDPPDNEs are able to provide improved ICG stability, binding specificity, and enhanced anticancer efficacy as compared to equal dosage of free ICG and/or DOX, showing a high potential for use in HER2 breast cancer therapy with reduced chemotoxicity. Electronic supplementary material The online version of this article (doi:10.1186/s12951-017-0274-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu-Hsiang Lee
- Department of Biomedical Sciences and Engineering, National Central University, No. 300, Jhongda Rd., Taoyuan City, 32001, Taiwan, ROC. .,Department of Chemical and Materials Engineering, National Central University, Taoyuan City, Taiwan, ROC.
| | - Yun-Ting Ma
- Department of Biomedical Sciences and Engineering, National Central University, No. 300, Jhongda Rd., Taoyuan City, 32001, Taiwan, ROC
| |
Collapse
|
26
|
Lee YH, Chang DS. Fabrication, characterization, and biological evaluation of anti-HER2 indocyanine green-doxorubicin-encapsulated PEG-b-PLGA copolymeric nanoparticles for targeted photochemotherapy of breast cancer cells. Sci Rep 2017; 7:46688. [PMID: 28429764 PMCID: PMC5399361 DOI: 10.1038/srep46688] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/24/2017] [Indexed: 01/24/2023] Open
Abstract
In this study, we aimed to develop anti-human epidermal growth factor receptor 2 (HER2) indocyanine green (ICG)-doxorubicin (DOX)-encapsulated polyethylene glycol-poly(lactic-co-glycolic acid) diblock copolymeric nanoparticles (HIDPPNPs) to explore the co-administration of phototherapy and chemotherapy for HER2-overexpressing breast cancer, a highly aggressive and medicine-resistant breast carcinoma. The HIDPPNPs were fabricated using a solvent evaporation technique followed by carbodiimide-mediated antibody conjugation on the nanoparticle surface. Compared with freely dissolved ICG, the HIDPPNPs conferred enhanced thermal stability to the entrapped ICG, were able to generate a hyperthermia effect at concentrations ≥1 μM ICG equivalent and provided increased production of singlet oxygen under 808-nm laser irradiation with an intensity of 6 W/cm2. Furthermore, the uptake efficiency of the HIDPPNPs in MDA-MB-453/HER2(+) cells was approximately 2-fold higher than that in MCF7/HER2(-) cells, demonstrating that the HIDPPNPs specifically target HER2-expressing cells. Based on the viability analysis, the HIDPPNPs exhibited effective cytotoxicity upon light exposure (808 nm; 6 W/cm2), and the resulting cell death rate was even higher than that caused by using twice amount of encapsulated DOX or ICG alone. These results indicate that the developed HIDPPNPs may serve as a feasible tool for use in anti-HER2 breast cancer therapy with reduced chemotoxicity.
Collapse
Affiliation(s)
- Yu-Hsiang Lee
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan R.O.C
- Department of Chemical and Materials Engineering, National Central University, Taoyuan City, Taiwan R.O.C
| | - Da-Sheng Chang
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan R.O.C
| |
Collapse
|
27
|
Zhang SK, Song JW, Li SB, Gao HW, Chang HY, Jia LL, Gong F, Tan YX, Ji SP. Design of pH-sensitive peptides from natural antimicrobial peptides for enhancing polyethylenimine-mediated gene transfection. J Gene Med 2017; 19. [PMID: 28370835 DOI: 10.1002/jgm.2955] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/30/2017] [Accepted: 03/30/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Poor endosomal release is a major barrier of polyplex-mediated gene transfection. Antimicrobial peptides (AMPs) are commonly used to improve polyethylenimine (PEI)-mediated gene transfection by increasing endosomal release. In the present study, we designed novel pH-sensitive peptides that highly enhance transfection efficiency compared to their parent peptides. METHODS Two analogues of melittin (Mel) and RV-23 (RV) were synthesized by replacing the positively-charged residues in their sequences with glutamic acid residues. The pH-sensitive lysis ability of the peptides, the effect of the peptides on physicochemical characteristics, the intracellular trafficking, the transfection efficiency, and the cytotoxicity of the polyplexes were determined. RESULTS The acidic peptides showed pH-sensitive lytic activity. The hemolytic activity of acidic peptides at pH 5.0 was higher than that at pH 7.4. The incorporation of acidic peptides did not affect the DNA binding ability of PEI but affected the physicochemical characteristics of the PEI/DNA polyplexes, which may be beneficial for endosomal release and gene transfection. The incorporation of acidic peptides into PEI/DNA polyplexes enhanced the PEI-mediated transfection efficiency corresponding to up to 42-fold higher luciferase activity compared to that of PEI alone. CONCLUSIONS The results of the present study indicate that replacement of positively-charged residues with glutamic acid residues in the AMP sequence yields pH-sensitive peptides, which enhance the transfection efficiency of PEI/DNA polyplexes in various cell lines.
Collapse
Affiliation(s)
- Shi-Kun Zhang
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Jin-Wen Song
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Su-Bo Li
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Hong-Wei Gao
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Hong-Yu Chang
- Department of Paediatrics, General Hospital of the PLA Rocket Force, Beijing, China
| | - Li-Li Jia
- Neonatal Department of Xi'an No 4 Hospital, Xi'an, China
| | - Feng Gong
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Ying-Xia Tan
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Shou-Ping Ji
- Department of Tissue Engineering, Beijing Institute of Transfusion Medicine, Beijing, China
| |
Collapse
|
28
|
Lee YH, Lai YH. Synthesis, Characterization, and Biological Evaluation of Anti-HER2 Indocyanine Green-Encapsulated PEG-Coated PLGA Nanoparticles for Targeted Phototherapy of Breast Cancer Cells. PLoS One 2016; 11:e0168192. [PMID: 27942034 PMCID: PMC5152925 DOI: 10.1371/journal.pone.0168192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2)-overexpressed breast cancer is known to be more aggressive and resistant to medicinal treatment and therefore to whom an alternative therapeutics is needed. Indocyanine green (ICG) has been widely exploited in breast cancer phototherapy. However, drawbacks of accelerated degradation and short half-life (2-4 min) in blood seriously hamper its use in the clinic. To overcome these challenges, an anti-HER2 ICG-encapsulated polyethylene glycol-coated poly(lactic-co-glycolic acid) nanoparticles (HIPPNPs) were developed in this study. Through the analyses of degradation rate coefficients of ICG with and without polymeric encapsulation, the photostability of HIPPNP-entrapped ICG significantly enhanced 4 folds (P < 0.05) while its thermal stabilities at 4 and 37°C significantly enhanced 5 and 3 (P < 0.05 for each) folds, respectively, under equal lighting and/or heating treatment for 48 h. The target specificity of HIPPNPs to HER2-positive cells was demonstrated based on a 6-fold (P < 0.05) enhancement of uptake efficiency of HIPPNPs in MDA-MB-453/HER2(+) cells within 4 h as compared with that in MCF7/HER2(-) cells. Moreover, the HIPPNPs with ≤ 25 μM ICG equivalent were nontoxic to cells in the absence of light illumination, and enabled to generate similar amount of singlet oxygen and hyperthermia effect as compared with that used by free ICG upon NIR irradiation. After 808 nm-laser irradiation with intensity of 6 W/cm2 for 5 min, the viability of MDA-MB-453 cells pre-treated by HIPPNPs with ≥ 5 μM ICG equivalent for 4 h significantly reduced as compared with that treated by equal concentration of free ICG (P < 0.05) and > 90% of the cells were eradicated while the dose of HIPPNPs was increased to 25 μM ICG equivalent. In summary, the developed HIPPNPs are anticipated as a feasible tool for use in phototherapy of breast cancer cells with HER2 expression.
Collapse
Affiliation(s)
- Yu-Hsiang Lee
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan R.O.C.
- Department of Chemical and Materials Engineering, National Central University, Taoyuan City, Taiwan R.O.C.
| | - Yun-Han Lai
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan R.O.C.
| |
Collapse
|
29
|
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: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
30
|
Berry S, Mastorakos P, Zhang C, Song E, Patel H, Suk JS, Hanes J. Enhancing Intracranial Delivery of Clinically Relevant Non-viral Gene Vectors. RSC Adv 2016; 48:41665-41674. [PMID: 27642512 DOI: 10.1039/c6ra01546h] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gene therapy is a promising strategy for the management of various neurological disorders that do not respond adequately to conventional therapeutics. The development of gene vectors with favorable safety profiles that can achieve uniform distribution and high-level transgene expression in the brain remains challenging. The rod-shaped, non-viral gene delivery platform based on poly-L-lysine (PLL) conjugated to a single segment of polyethylene glycol (PEG) has shown safe transfection in human nares and mouse brains in vivo. However, we have previously demonstrated that a denser PEG coating is required for rapid diffusion of nanoparticles in the brain extracellular space. Here, we engineered a densely PEGylated version of this platform based on PLL polymers conjugated to branched PEG via alkyne-azide cycloaddition. We found that the newly developed gene vectors rapidly diffused in the brain parenchyma, providing significantly improved vector distribution and overall transgene expression in vivo compared to the previously developed platform. These brain-penetrating DNA nanoparticles exhibited enhanced cellular uptake presumably due to their ellipsoidal morphology. By simultaneously improving delivery to target cells and subsequent transfection, our densely PEGylated PLL DNA nanoparticles can provide widespread, high levels of transgene expression, essential for effective targeting of highly disseminated brain diseases.
Collapse
Affiliation(s)
- Sneha Berry
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Panagiotis Mastorakos
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clark Zhang
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric Song
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Himat Patel
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jung Soo Suk
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Justin Hanes
- The Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
31
|
Sanada Y, Shiomi T, Okobira T, Tan M, Nishikawa M, Akiba I, Takakura Y, Sakurai K. Polypod-Shaped DNAs: Small-Angle X-ray Scattering and Immunostimulatory Activity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:3760-3765. [PMID: 27007061 DOI: 10.1021/acs.langmuir.6b00398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
We explored in detail the relationship between the structure in aqueous solution and immunostimulatory activity of polypod-shaped DNAs, called polypodnas. The polypodnas were constructed using 3-6 oligodeoxynucleotides (ODNs) to obtain tri-, tetra-, penta-, and hexapodna, each of which had 3, 4, 5, and 6 arms made of double-stranded DNA, respectively. A highly potent immunostimulatory CpG sequence was included into each of the polypodnas. Synchrotron X-ray scattering analysis showed that the double-stranded DNA arms of all of the polypodnas adopted a B-form DNA conformation. The analysis also suggested that some nucleotides in the central parts of pentapodna and hexapodna did not form base pairs, whereas those of tripodna and tetrapodna all formed base pairs. This difference would occur because of an increase in steric hindrance and electrical repulsion with increasing number of arms. The pentapodna and hexapodna induced a large amount of tumor necrosis factor α-release from macrophage-like cells compared with the tripodna and tetrapodna, suggesting that the partly loosened DNA in polypodna with many arms is advantageous for exposing the immunostimulatory sequences of the polypodna.
Collapse
Affiliation(s)
- Yusuke Sanada
- Department of Chemistry and Biochemistry, University of Kitakyushu , 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
- Structural Materials Science Laboratory SPring-8 Center, RIKEN Harima Institute Research 1-1-1 Kouto, Sayo-cho, Sayo, Hyogo 679-5148, Japan
| | - Tomoki Shiomi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University , 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tadashi Okobira
- Department of Chemical Science and Engineering, Ariake National College of Technology , 150 Higashihagio-Machi, Omuta, Fukuoka 836-8585, Japan
| | - Mengmeng Tan
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University , 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Makiya Nishikawa
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University , 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Isamu Akiba
- Department of Chemistry and Biochemistry, University of Kitakyushu , 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
- Structural Materials Science Laboratory SPring-8 Center, RIKEN Harima Institute Research 1-1-1 Kouto, Sayo-cho, Sayo, Hyogo 679-5148, Japan
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University , 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuo Sakurai
- Department of Chemistry and Biochemistry, University of Kitakyushu , 1-1 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
- Structural Materials Science Laboratory SPring-8 Center, RIKEN Harima Institute Research 1-1-1 Kouto, Sayo-cho, Sayo, Hyogo 679-5148, Japan
| |
Collapse
|
32
|
Chen W, Li H, Liu Z, Yuan W. Lipopolyplex for Therapeutic Gene Delivery and Its Application for the Treatment of Parkinson's Disease. Front Aging Neurosci 2016; 8:68. [PMID: 27092073 PMCID: PMC4820442 DOI: 10.3389/fnagi.2016.00068] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/21/2016] [Indexed: 01/10/2023] Open
Abstract
Lipopolyplex is a core-shell structure composed of nucleic acid, polycation and lipid. As a non-viral gene delivery vector, lipopolyplex combining the advantages of polyplex and lipoplex has shown superior colloidal stability, reduced cytotoxicity, extremely high gene transfection efficiency. Following intravenous administration, there are many strategies based on lipopolyplex to overcome the complex biological barriers in systemic gene delivery including condensation of nucleic acids into nanoparticles, long circulation, cell targeting, endosomal escape, release to cytoplasm and entry into cell nucleus. Parkinson's disease (PD) is the second most common neurodegenerative disorder and severely influences the patients' life quality. Current gene therapy clinical trials for PD employing viral vectors didn't achieve satisfactory efficacy. However, lipopolyplex may become a promising alternative approach owing to its stability in blood, ability to cross the blood-brain barrier (BBB) and specific targeting to diseased brain cells.
Collapse
Affiliation(s)
- Wei Chen
- Department of Neurology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine Shanghai, China
| | - Hui Li
- School of Pharmacy, Shanghai JiaoTong University Shanghai, China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine Shanghai, China
| | - Weien Yuan
- School of Pharmacy, Shanghai JiaoTong University Shanghai, China
| |
Collapse
|
33
|
Rheiner S, Bae Y. Increased poly(ethylene glycol) density decreases transfection efficacy of siRNA/poly(ethylene imine) complexes. AIMS BIOENGINEERING 2016. [DOI: 10.3934/bioeng.2016.4.454] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
34
|
Bishop CJ, Kozielski KL, Green JJ. Exploring the role of polymer structure on intracellular nucleic acid delivery via polymeric nanoparticles. J Control Release 2015; 219:488-499. [PMID: 26433125 DOI: 10.1016/j.jconrel.2015.09.046] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 11/08/2022]
Abstract
Intracellular nucleic acid delivery has the potential to treat many genetically-based diseases, however, gene delivery safety and efficacy remains a challenging obstacle. One promising approach is the use of polymers to form polymeric nanoparticles with nucleic acids that have led to exciting advances in non-viral gene delivery. Understanding the successes and failures of gene delivery polymers and structures is the key to engineering optimal polymers for gene delivery in the future. This article discusses the polymer structural features that enable effective intracellular delivery of DNA and RNA, including protection of nucleic acid cargo, cellular uptake, endosomal escape, vector unpacking, and delivery to the intracellular site of activity. The chemical properties that aid in each step of intracellular nucleic acid delivery are described and specific structures of note are highlighted. Understanding the chemical design parameters of polymeric nucleic acid delivery nanoparticles is important to achieving the goal of safe and effective non-viral genetic nanomedicine.
Collapse
Affiliation(s)
- Corey J Bishop
- Department of Biomedical Engineering, Institute for Nanobiotechnology, Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kristen L Kozielski
- Department of Biomedical Engineering, Institute for Nanobiotechnology, Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jordan J Green
- Department of Biomedical Engineering, Institute for Nanobiotechnology, Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Departments of Neurosurgery, Oncology, and Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
35
|
Okuda T, Suzuki Y, Kobayashi Y, Ishii T, Uchida S, Itaka K, Kataoka K, Okamoto H. Development of Biodegradable Polycation-Based Inhalable Dry Gene Powders by Spray Freeze Drying. Pharmaceutics 2015; 7:233-54. [PMID: 26343708 PMCID: PMC4588198 DOI: 10.3390/pharmaceutics7030233] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 12/27/2022] Open
Abstract
In this study, two types of biodegradable polycation (PAsp(DET) homopolymer and PEG-PAsp(DET) copolymer) were applied as vectors for inhalable dry gene powders prepared by spray freeze drying (SFD). The prepared dry gene powders had spherical and porous structures with a 5~10-μm diameter, and the integrity of plasmid DNA could be maintained during powder production. Furthermore, it was clarified that PEG-PAsp(DET)-based dry gene powder could more sufficiently maintain both the physicochemical properties and in vitro gene transfection efficiencies of polyplexes reconstituted after powder production than PAsp(DET)-based dry gene powder. From an in vitro inhalation study using an Andersen cascade impactor, it was demonstrated that the addition of l-leucine could markedly improve the inhalation performance of dry powders prepared by SFD. Following pulmonary delivery to mice, both PAsp(DET)- and PEG-PAsp(DET)-based dry gene powders could achieve higher gene transfection efficiencies in the lungs compared with a chitosan-based dry gene powder previously reported by us.
Collapse
Affiliation(s)
- Tomoyuki Okuda
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan.
| | - Yumiko Suzuki
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan.
| | - Yuko Kobayashi
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan.
| | - Takehiko Ishii
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo 113-8656, Japan.
| | - Satoshi Uchida
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Keiji Itaka
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Kazunori Kataoka
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo 113-8656, Japan.
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Hirokazu Okamoto
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan.
| |
Collapse
|
36
|
Sandoval S, Mendez N, Alfaro JG, Yang J, Aschemeyer S, Liberman A, Trogler WC, Kummel AC. Quantification of endocytosis using a folate functionalized silica hollow nanoshell platform. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:88003. [PMID: 26315280 PMCID: PMC5996829 DOI: 10.1117/1.jbo.20.8.088003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/27/2015] [Indexed: 06/04/2023]
Abstract
A quantification method to measure endocytosis was designed to assess cellular uptake and specificity of a targeting nanoparticle platform. A simple N -hydroxysuccinimide ester conjugation technique to functionalize 100-nm hollow silica nanoshell particles with fluorescent reporter fluorescein isothiocyanate and folate or polyethylene glycol (PEG) was developed. Functionalized nanoshells were characterized using scanning electron microscopy and transmission electron microscopy and the maximum amount of folate functionalized on nanoshell surfaces was quantified with UV-Vis spectroscopy. The extent of endocytosis by HeLa cervical cancer cells and human foreskin fibroblast (HFF-1) cells was investigated in vitro using fluorescence and confocal microscopy. A simple fluorescence ratio analysis was developed to quantify endocytosis versus surface adhesion. Nanoshells functionalized with folate showed enhanced endocytosis by cancer cells when compared to PEG functionalized nanoshells. Fluorescence ratio analyses showed that 95% of folate functionalized silica nanoshells which adhered to cancer cells were endocytosed, while only 27% of PEG functionalized nanoshells adhered to the cell surface and underwent endocytosis when functionalized with 200 and 900 μg , respectively. Additionally, the endocytosis of folate functionalized nanoshells proved to be cancer cell selective while sparing normal cells. The developed fluorescence ratio analysis is a simple and rapid verification/validation method to quantify cellular uptake between datasets by using an internal control for normalization.
Collapse
Affiliation(s)
- Sergio Sandoval
- University of California, San Diego, Moores Cancer Center, Department of Bioengineering, CalIT Nanomedicine Laboratory, La Jolla, California 92093, United States
| | - Natalie Mendez
- University of California, San Diego, Department of Nanoengineering, Chemical Engineering, and Material Science, La Jolla, California 92093, United States
| | - Jesus G. Alfaro
- University of California, San Diego, Department of Nanoengineering, Chemical Engineering, and Material Science, La Jolla, California 92093, United States
| | - Jian Yang
- University of California, San Diego, Department of Nanoengineering, Chemical Engineering, and Material Science, La Jolla, California 92093, United States
| | - Sharraya Aschemeyer
- University of California, San Diego, Department of Chemistry and Biochemistry, La Jolla, California 92093, United States
| | - Alex Liberman
- University of California, San Diego, Department of Nanoengineering, Chemical Engineering, and Material Science, La Jolla, California 92093, United States
| | - William C. Trogler
- University of California, San Diego, Department of Chemistry and Biochemistry, La Jolla, California 92093, United States
| | - Andrew C. Kummel
- University of California, San Diego, Department of Chemistry and Biochemistry, La Jolla, California 92093, United States
| |
Collapse
|
37
|
Mastorakos P, da Silva AL, Chisholm J, Song E, Choi WK, Boyle MP, Morales MM, Hanes J, Suk JS. Highly compacted biodegradable DNA nanoparticles capable of overcoming the mucus barrier for inhaled lung gene therapy. Proc Natl Acad Sci U S A 2015; 112:8720-5. [PMID: 26124127 PMCID: PMC4507234 DOI: 10.1073/pnas.1502281112] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gene therapy has emerged as an alternative for the treatment of diseases refractory to conventional therapeutics. Synthetic nanoparticle-based gene delivery systems offer highly tunable platforms for the delivery of therapeutic genes. However, the inability to achieve sustained, high-level transgene expression in vivo presents a significant hurdle. The respiratory system, although readily accessible, remains a challenging target, as effective gene therapy mandates colloidal stability in physiological fluids and the ability to overcome biological barriers found in the lung. We formulated highly stable DNA nanoparticles based on state-of-the-art biodegradable polymers, poly(β-amino esters) (PBAEs), possessing a dense corona of polyethylene glycol. We found that these nanoparticles efficiently penetrated the nanoporous and highly adhesive human mucus gel layer that constitutes a primary barrier to reaching the underlying epithelium. We also discovered that these PBAE-based mucus-penetrating DNA nanoparticles (PBAE-MPPs) provided uniform and high-level transgene expression throughout the mouse lungs, superior to several gold standard gene delivery systems. PBAE-MPPs achieved robust transgene expression over at least 4 mo following a single administration, and their transfection efficiency was not attenuated by repeated administrations, underscoring their clinical relevance. Importantly, PBAE-MPPs demonstrated a favorable safety profile with no signs of toxicity following intratracheal administration.
Collapse
Affiliation(s)
- Panagiotis Mastorakos
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21297
| | - Adriana L da Silva
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941902, Brazil
| | - Jane Chisholm
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Eric Song
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231; Center for Biotechnology Education, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218
| | - Won Kyu Choi
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Michael P Boyle
- Adult Cystic Fibrosis Program, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Marcelo M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941902, Brazil
| | - Justin Hanes
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21297; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218;
| | - Jung Soo Suk
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21297;
| |
Collapse
|
38
|
Leng Q, Chou ST, Scaria PV, Woodle MC, Mixson AJ. Increased tumor distribution and expression of histidine-rich plasmid polyplexes. J Gene Med 2015; 16:317-28. [PMID: 25303767 PMCID: PMC4242722 DOI: 10.1002/jgm.2807] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/10/2014] [Indexed: 12/12/2022] Open
Abstract
Background Selecting nonviral carriers for in vivo gene delivery is often dependent on determining the optimal carriers from transfection assays in vitro. The rationale behind this in vitro strategy is to cast a net sufficiently wide to identify the few effective carriers of plasmids for in vivo studies. Nevertheless, many effective in vivo carriers may be overlooked by this strategy because of the marked differences between in vitro and in vivo assays. Methods After solid-phase synthesis of linear and branched histidine/lysine (HK) peptides, the two peptide carriers were compared for their ability to transfect MDA-MB-435 tumor cells in vitro and then in vivo. Results By contrast to their transfection activity in vitro, the linear H2K carrier of plasmids was far more effective in vivo compared to the branch H2K4b. Surprisingly, negatively-charged polyplexes formed by the linear H2K peptide gave higher transfection in vivo than did those with a positive surface charge. To examine the distribution of plasmid expression within the tumor from H2K polyplexes, we found widespread expression by immunohistochemical staining. With a fluorescent tdTomato expressing-plasmid, we confirmed a pervasive distribution and gene expression within the tumor mediated by the H2K polyplex. Conclusions Although mechanisms underlying the efficiency of gene expression are probably multifactorial, unpacking of the H2K polyplex within the tumor appears to have a significant role. Further development of these H2K polyplexes represents an attractive approach for plasmid-based therapies of cancer. © 2014 The Authors. The Journal of Gene Medicine published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Qixin Leng
- Department of Pathology, University Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
39
|
Liao R, Yi S, Liu M, Jin W, Yang B. Folic-Acid-Targeted Self-Assembling Supramolecular Carrier for Gene Delivery. Chembiochem 2015; 16:1622-8. [DOI: 10.1002/cbic.201500208] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Indexed: 12/27/2022]
|
40
|
Mastorakos P, Zhang C, Berry S, Oh Y, Lee S, Eberhart CG, Woodworth GF, Suk JS, Hanes J. Highly PEGylated DNA Nanoparticles Provide Uniform and Widespread Gene Transfer in the Brain. Adv Healthc Mater 2015; 4:1023-33. [PMID: 25761435 DOI: 10.1002/adhm.201400800] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/15/2015] [Indexed: 12/13/2022]
Abstract
Gene delivery to the central nervous system (CNS) has potential as a means for treating numerous debilitating neurological diseases. Nonviral gene vector platforms are tailorable and can overcome key limitations intrinsic to virus-mediated delivery; however, lack of clinical efficacy with nonviral systems to date may be attributed to limited gene vector dispersion and transfection in vivo. It is shown that the brain extracellular matrix (ECM) strongly limits penetration of polymer-based gene vector nanoparticles (NP) through the brain parenchyma, even when they are very small (<60 nm) and coated with a polyethylene glycol (PEG) corona of typical density. Following convection enhanced delivery (CED), conventional gene vectors are confined to the injection site, presumably by adhesive interactions with the brain ECM and do not provide gene expression beyond the point of administration. In contrast, it is found that incorporating highly PEGylated polymers allows the production of compacted (≈43 nm) and colloidally stable DNA NP that avoid adhesive trapping within the brain parenchyma. When administered by CED into the rat striatum, highly PEGylated DNA NP distribute throughout and provide broad transgene expression without vector-induced toxicity. The use of these brain-penetrating gene vectors, in conjunction with CED, offers an avenue to improve gene therapy for CNS diseases.
Collapse
Affiliation(s)
- Panagiotis Mastorakos
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Department of Ophthalmology; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 600 N. Wolfe Street Baltimore MD 21297 USA
| | - Clark Zhang
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Department of Biomedical Engineering; Johns Hopkins University School of Medicine; 720 Rutland Avenue Baltimore MD 21205 USA
| | - Sneha Berry
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Center for Biotechnology Education; Krieger School of Arts and Sciences; Johns Hopkins University; 3400 N. Charles Street Baltimore MD 21218 USA
| | - Yumin Oh
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Russell H. Morgan Department of Radiology and Radiological Science; Johns Hopkins University; 601 N. Caroline Street Baltimore MD 21287 USA
| | - Seulki Lee
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Russell H. Morgan Department of Radiology and Radiological Science; Johns Hopkins University; 601 N. Caroline Street Baltimore MD 21287 USA
| | - Charles G. Eberhart
- Department of Pathology; Johns Hopkins University School of Medicine; 600 N. Wolfe Street Baltimore MD 21287 USA
| | - Graeme F. Woodworth
- Departments of Neurological Surgery Anatomy and Neurobiology; University of Maryland School of Medicine; 22 S. Greene Street Baltimore MD 21201 USA
| | - Jung Soo Suk
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Department of Ophthalmology; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 600 N. Wolfe Street Baltimore MD 21297 USA
| | - Justin Hanes
- Center for Nanomedicine; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 400 N. Broadway Baltimore MD 21231 USA
- Department of Ophthalmology; The Wilmer Eye Institute; Johns Hopkins University School of Medicine; 600 N. Wolfe Street Baltimore MD 21297 USA
- Department of Biomedical Engineering; Johns Hopkins University School of Medicine; 720 Rutland Avenue Baltimore MD 21205 USA
- Department of Chemical and Biomolecular Engineering; Johns Hopkins University; 3400 N. Charles Street Baltimore MD 21218 USA
| |
Collapse
|
41
|
Enhanced gene silencing through human serum albumin-mediated delivery of polyethylenimine-siRNA polyplexes. PLoS One 2015; 10:e0122581. [PMID: 25856158 PMCID: PMC4391875 DOI: 10.1371/journal.pone.0122581] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 02/17/2015] [Indexed: 11/19/2022] Open
Abstract
Small interfering RNA (siRNA) targeted therapeutics (STT) offers a compelling alternative to tradition medications for treatment of genetic diseases by providing a means to silence the expression of specific aberrant proteins, through interference at the expression level. The perceived advantage of siRNA therapy is its ability to target, through synthetic antisense oligonucleotides, any part of the genome. Although STT provides a high level of specificity, it is also hindered by poor intracellular uptake, limited blood stability, high degradability and non-specific immune stimulation. Since serum proteins has been considered as useful vehicles for targeting tumors, in this study we investigated the effect of incorporation of human serum albumin (HSA) in branched polyethylenimine (bPEI)-siRNA polyplexes in their internalization in epithelial and endothelial cells. We observed that introduction of HSA preserves the capacity of bPEI to complex with siRNA and protect it against extracellular endonucleases, while affording significantly improved internalization and silencing efficiency, compared to bPEI-siRNA polyplexes in endothelial and metastatic breast cancer epithelial cells. Furthermore, the uptake of the HSA-bPEI-siRNA ternary polyplexes occurred primarily through a caveolae-mediated endocytosis, thus providing evidence for a clear role for HSA in polyplex internalization. These results provide further impetus to explore the role of serum proteins in delivery of siRNA.
Collapse
|
42
|
Foster AA, Ross NL, Sullivan MO. Fluorescent dye incorporation causes weakened gene association and intracellular aggregate formation in nonviral carriers. J Gene Med 2015; 17:69-79. [PMID: 25731756 DOI: 10.1002/jgm.2824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 02/14/2015] [Accepted: 02/26/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The successful application of nonviral gene transfer technologies requires both improved understanding and control with respect to intracellular trafficking and release. However, the intracellular space is highly complex and hence well-defined, stable structures are necessary to probe the stages of the delivery pathway. Fluorescent labeling is a regularly used approach to monitor nonviral delivery and release, yet few studies investigate the effects of label incorporation on the structure and activity of gene-containing vehicles. METHODS In the present study, the impacts of label incorporation on the assembly and gene transfer capacity of DNA polyplexes were determined through the utilization of a model DNA-polyethylenimine (PEI) delivery system. PEI was fluorescently labeled with the Oregon Green® dye prior to polyplex formation and delivery to CHO-K1 cells. RESULTS The present study provides evidence showing that routine labeling strategies for polyplexes weakened DNA binding affinity, produced large quantities of extracellular structures and significantly increased intracellular polyplex aggregation. Additionally, cellular internalization studies showed that increased labeling fractions led to reductions in polyplex uptake as a result of weakened complexation. CONCLUSIONS These results not only provide insight into the assembly of these structures, but also help to identify labeling strategies sufficient to preserve activity at the same time as enabling detailed studies of trafficking and disassembly.
Collapse
Affiliation(s)
- Abbygail A Foster
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Nikki L Ross
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| |
Collapse
|
43
|
Martin TM, Wysocki BJ, Wysocki TA, Pannier AK. Identifying Intracellular pDNA Losses From a Model of Nonviral Gene Delivery. IEEE Trans Nanobioscience 2015; 14:455-464. [PMID: 25622323 DOI: 10.1109/tnb.2015.2392777] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nonviral gene delivery systems are a type of nanocommunication system that transmit plasmid packets (i.e., pDNA packets) that are programmed at the nanoscale to biological systems at the microscopic cellular level. This engineered nanocommunication system suffers large pDNA losses during transmission of the genetically encoded information, preventing its use in biotechnological and medical applications. The pDNA losses largely remain uncharacterized, and the ramifications of reducing pDNA loss from newly designed gene delivery systems remain difficult to predict. Here, the pDNA losses during primary and secondary transmission chains were identified utilizing a MATLAB model employing queuing theory simulating delivery of pEGFPLuc transgene to HeLa cells carried by Lipofectamine 2000 nonviral DNA carrier. Minimizing pDNA loss during endosomal escape of the primary transmission process results in increased number of pDNA in the nucleus with increased transfection, but with increased probability of cell death. The number of pDNA copies in the nucleus and the amount of time the pDNAs are in the nucleus directly correlates to improved transfection efficiency. During secondary transmission, pDNAs are degraded during distribution to daughter cells. Reducing pDNA losses improves transfection, but a balance in quantity of nuclear pDNA, mitosis, and toxicity must be considered in order to achieve therapeutically relevant transfection levels.
Collapse
|
44
|
Prasannan A, Debele TA, Tsai HC, Chao CC, Lin CP, Hsiue GH. Synthesis and evaluation of the targeted binding of RGD-containing PEGylated-PEI/DNA polyplex micelles as radiotracers for a tumor-targeting imaging probe. RSC Adv 2015. [DOI: 10.1039/c5ra18644g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Polyplex micelles with pEGFP and RGD-modified poly(ethylene glycol)-grafted polyethylenimine (E[c(RGDyK)]2-PEG-g-PEI) and were labeled with 99mTc for the in vivo study as proficient probes for molecular imaging.
Collapse
Affiliation(s)
- Adhimoorthy Prasannan
- Graduate Institute of Applied Science and Technology
- National Taiwan University of Science and Technology
- Taipei 106
- Republic of China
- Department of Chemical Engineering/R&D Center for Membrane Technology
| | - Tilahun Ayane Debele
- Graduate Institute of Applied Science and Technology
- National Taiwan University of Science and Technology
- Taipei 106
- Republic of China
| | - Hsieh-Chih Tsai
- Graduate Institute of Applied Science and Technology
- National Taiwan University of Science and Technology
- Taipei 106
- Republic of China
| | - Chiz-Cheng Chao
- Department of Chemical Engineering
- National Chung Hsing University
- Taichung
- Republic of China
| | - Che-Ping Lin
- Department of Chemical Engineering
- National Tsing Hua University
- Hsinchu
- Republic of China
| | - Ging-Ho Hsiue
- Department of Chemical Engineering
- National Tsing Hua University
- Hsinchu
- Republic of China
- Department of Chemical Engineering/R&D Center for Membrane Technology
| |
Collapse
|
45
|
Dong H, Parekh HS, Xu ZP. Particle size- and number-dependent delivery to cells by layered double hydroxide nanoparticles. J Colloid Interface Sci 2015; 437:10-16. [DOI: 10.1016/j.jcis.2014.09.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/02/2014] [Accepted: 09/08/2014] [Indexed: 12/16/2022]
|
46
|
Hwang ME, Keswani RK, Pack DW. Dependence of PEI and PAMAM Gene Delivery on Clathrin- and Caveolin-Dependent Trafficking Pathways. Pharm Res 2014; 32:2051-9. [PMID: 25511918 DOI: 10.1007/s11095-014-1598-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/05/2014] [Indexed: 11/29/2022]
Abstract
PURPOSE Non-viral gene delivery vehicles such as polyethylenimine and polyamidoamine dendrimer effectively condense plasmid DNA, facilitate endocytosis, and deliver nucleic acid cargo to the nucleus in vitro. Better understanding of intracellular trafficking mechanisms involved in polymeric gene delivery is a prerequisite to clinical application. This study investigates the role of clathrin and caveolin endocytic pathways in cellular uptake and subsequent vector processing. METHODS We formed 25-kD polyethylenimine (PEI) and generation 4 (G4) polyamidoamine (PAMAM) polyplexes at N/P 10 and evaluated internalization pathways and gene delivery in HeLa cells. Clathrin- and caveolin-dependent endocytosis inhibitors were used at varying concentrations to elucidate the roles of these important pathways. RESULTS PEI and PAMAM polyplexes were internalized by both pathways. However, the amount of polyplex internalized poorly correlated with transgene expression. While the caveolin-dependent pathway generally led to effective gene delivery with both polymers, complete inhibition of the clathrin-dependent pathway was also deleterious to transfection with PEI polyplexes. Inhibition of one endocytic pathway may lead to an overall increase in uptake via unaffected pathways, suggesting the existence of compensatory endocytic mechanisms. CONCLUSIONS The well-studied clathrin- and caveolin-dependent endocytosis pathways are not necessarily independent, and perturbing one mechanism of trafficking influences the larger trafficking network.
Collapse
Affiliation(s)
- Mark E Hwang
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, 61801, USA
| | | | | |
Collapse
|
47
|
Dong H, Parekh HS, Xu ZP. Enhanced cellular delivery and biocompatibility of a small layered double hydroxide-liposome composite system. Pharmaceutics 2014; 6:584-98. [PMID: 25431895 PMCID: PMC4279134 DOI: 10.3390/pharmaceutics6040584] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 01/13/2023] Open
Abstract
The various classes of gene delivery vectors possess distinct advantages and disadvantages, each of which impacts on cargo loading, delivery and, ultimately, its function. With this in mind, herein we report on a small layered double hydroxide (sLDH)–liposome composite system, drawing upon the salient features of LDH and liposome classes of vectors, while avoiding their inherent shortfalls when used independently. sLDH–liposome composites were prepared by the hydration of freeze-dried matrix method. These composite systems, with a Z-average size of ≈200 nm, exhibited low cytotoxicity and demonstrated good suspension stability, both in water and cell culture medium after rehydration. Our studies demonstrate that short dsDNAs/ssDNAs were completely bound and protected in the composite system at an sLDH:DNA mass ratio of 20:1, regardless of the approach to DNA loading. This composite system delivered DNA to HCT-116 cells with ≈3-fold greater efficiency, when compared to sLDH alone. Our findings point towards the sLDH-liposome composite system being an effective and biocompatible gene delivery system.
Collapse
Affiliation(s)
- Haiyan Dong
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Harendra S Parekh
- Pharmacy Australia Centre of Excellence, School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| | - Zhi Ping Xu
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
48
|
Zhou Y, Zhang C, Liang W. Development of RNAi technology for targeted therapy — A track of siRNA based agents to RNAi therapeutics. J Control Release 2014; 193:270-81. [DOI: 10.1016/j.jconrel.2014.04.044] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 12/31/2022]
|
49
|
Smolny M, Rogers ML, Shafton A, Rush RA, Stebbing MJ. Development of non-viral vehicles for targeted gene transfer into microglia via the integrin receptor CD11b. Front Mol Neurosci 2014; 7:79. [PMID: 25346658 PMCID: PMC4191133 DOI: 10.3389/fnmol.2014.00079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 09/16/2014] [Indexed: 12/12/2022] Open
Abstract
Microglial activation is a central event in neurodegeneration. Novel technologies are sought for that specifically manipulate microglial function in order to delineate their role in onset and progression of neuropathologies. We investigated for the first time whether non-viral gene delivery based on polyethyleneglycol-polyethyleneimine conjugated to the monoclonal anti-CD11b antibody OX42 ("OX42-immunogene") could be used to specifically target microglia. We first conducted immunofluorescence studies with the OX42 antibody and identified its microglial integrin receptor CD11b as a potential target for receptor-mediated gene transfer based on its cellular specificity in mixed glia culture and in vivo and found that the OX42 antibody is rapidly internalized and trafficked to acidic organelles in absence of activation of the respiratory burst. We then performed transfection experiments with the OX42-immunogene in vitro and in rat brain showing that the OX42-immunogene although internalized was degraded intracellularly and did not cause substantial gene expression in microglia. Investigation of specific barriers to microglial gene transfer revealed that aggregated OX42-immunogene polyplexes stimulated the respiratory burst that likely involved Fcγ-receptors. Transfections in the presence of the endosomolytic agent chloroquine improved transfection efficiency indicating that endosomal escape may be limited. This study identifies CD11b as an entry point for antibody-mediated gene transfer into microglia and takes important steps toward the further development of OX42-immunogenes.
Collapse
Affiliation(s)
- Markus Smolny
- School of Medical Sciences and Health Innovations Research Institute, Royal Melbourne Institute of Technology UniversityBundoora, VIC, Australia
| | - Mary-Louise Rogers
- Department of Human Physiology, Centre for Neuroscience, Flinders UniversityAdelaide, SA, Australia
| | - Anthony Shafton
- The Florey Institute of Neuroscience and Mental Health, The University of MelbourneParkville, VIC, Australia
| | - Robert A. Rush
- Department of Human Physiology, Centre for Neuroscience, Flinders UniversityAdelaide, SA, Australia
| | - Martin J. Stebbing
- School of Medical Sciences and Health Innovations Research Institute, Royal Melbourne Institute of Technology UniversityBundoora, VIC, Australia
| |
Collapse
|
50
|
Abstract
The use of nanoparticulate pharmaceutical drug delivery systems (NDDSs) to enhance the in vivo effectiveness of drugs is now well established. The development of multifunctional and stimulus-sensitive NDDSs is an active area of current research. Such NDDSs can have long circulation times, target the site of the disease and enhance the intracellular delivery of a drug. This type of NDDS can also respond to local stimuli that are characteristic of the pathological site by, for example, releasing an entrapped drug or shedding a protective coating, thus facilitating the interaction between drug-loaded nanocarriers and target cells or tissues. In addition, imaging contrast moieties can be attached to these carriers to track their real-time biodistribution and accumulation in target cells or tissues. Here, I highlight recent developments with multifunctional and stimuli-sensitive NDDSs and their therapeutic potential for diseases including cancer, cardiovascular diseases and infectious diseases.
Collapse
|