1
|
Song BF, Li BJ, Sun Y, Li M, Rao T, Ruan Y, Cheng F. GOLPH3 promotes calcium oxalate-induced renal injury and fibrosis through Golgi stress-mediated apoptosis and inflammatory responses. Sci Rep 2025; 15:7640. [PMID: 40038402 PMCID: PMC11880244 DOI: 10.1038/s41598-025-91638-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
A common urological disorder, calcium oxalate (CaOx) stones are the most common form of kidney stones. Deposition of CaOx crystals leads to tubular damage, interstitial fibrosis, and chronic kidney disease. Understanding the intrinsic mechanisms of kidney stone formation is essential for the prevention of kidney stones and the development of new therapeutic agents. The Golgi apparatus is a key organelle in the secretory pathway of eukaryotic cells, which plays an important role in the sorting, modification, and transport of proteins within the cell, and has been reported to be involved in several diseases, including prostate tumors, gastrointestinal tumors, sepsis, and so on. GOLPH3 is also known as GPP34, GMx33, or MIDAS. It is a glycoprotein that regulates traffic between the trans-Golgi network and the cell membrane. However, its role in renal injury caused by CaOx crystal deposition is still unclear. Results from immunohistochemistry, qRT-PCR, western blot, and public database single nucleotide RNA-seq showed that GOLPH3 was significantly upregulated in kidney stone patients and animal kidneys. Significant inhibition of Golgi stress, apoptosis, and renal fibrosis by GOLPH3 inhibition with siRNA in CaOx-stimulated HK-2 cells. The PI3K\AKT\mTOR signaling pathway was inhibited by GOLPH3 knockdown, which may be associated with reduced inflammatory response and apoptosis, as well as restoration of Golgi morphology and function. In conclusion, GOLPH3 plays a critical role in CaOx-induced kidney injury by promoting Golgi stress and increasing inflammatory responses, apoptosis, and renal fibrosis, suggesting that GOLPH3 is a potential therapeutic target for kidney stones.
Collapse
Affiliation(s)
- Bao-Feng Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo-Jun Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yushi Sun
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ming Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yuan Ruan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
2
|
Biscaia-Caleiras M, Fonseca NA, Lourenço AS, Moreira JN, Simões S. Rational formulation and industrial manufacturing of lipid-based complex injectables: Landmarks and trends. J Control Release 2024; 373:617-639. [PMID: 39002799 DOI: 10.1016/j.jconrel.2024.07.021] [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: 04/05/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Lipid-based complex injectables are renowned for their effectiveness in delivering drugs, with many approved products. While significant strides have been made in formulating nanosystems for small molecular weight drugs, a pivotal breakthrough emerged with the recognition of lipid nanoparticles as a promising platform for delivering nucleic acids. This finding has paved the way for tackling long-standing challenges in molecular and delivery aspects (e.g., mRNA stability, intracellular delivery) that have impeded the clinical translation of gene therapy, especially in the realm of immunotherapy. Nonetheless, developing and implementing new lipid-based delivery systems pose significant challenges, as industrial manufacturing of these formulations often involves complex, multi-batch processes, giving rise to issues related to scalability, stability, sterility, and regulatory compliance. To overcome these obstacles, embracing the principles of quality-by-design (QbD) is imperative. Furthermore, adopting cutting-edge manufacturing and process analytical tools (PAT) that facilitate the transition from batch to continuous production is essential. Herein, the key milestones and insights derived from the development of currently approved lipid- nanosystems will be explored. Additionally, a comprehensive and critical overview of the latest technologies and regulatory guidelines that underpin the creation of more efficient, scalable, and flexible manufacturing processes for complex lipid-based nanoformulations will be provided.
Collapse
Affiliation(s)
- Mariana Biscaia-Caleiras
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal; Univ Coimbra-University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Nuno A Fonseca
- Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal
| | - Ana Sofia Lourenço
- Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal
| | - João Nuno Moreira
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; Univ Coimbra-University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Sérgio Simões
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; Bluepharma-Indústria Farmacêutica, S.A., São Martinho do Bispo, 3045-016 Coimbra, Portugal; Univ Coimbra-University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
3
|
Abdellatif AAH, Bouazzaoui A, Tawfeek HM, Younis MA. MCT4 knockdown by tumor microenvironment-responsive nanoparticles remodels the cytokine profile and eradicates aggressive breast cancer cells. Colloids Surf B Biointerfaces 2024; 238:113930. [PMID: 38692174 DOI: 10.1016/j.colsurfb.2024.113930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
Breast cancer is a wide-spread threat to the women's health. The drawbacks of conventional treatments necessitate the development of alternative strategies, where gene therapy has regained hope in achieving an efficient eradication of aggressive tumors. Monocarboxylate transporter 4 (MCT4) plays pivotal roles in the growth and survival of various tumors, which offers a promising target for treatment. In the present study, pH-responsive lipid nanoparticles (LNPs) based on the ionizable lipid,1,2-dioleoyl-3-dimethylammonium propane (DODAP), were designed for the delivery of siRNA targeting MCT4 gene to the breast cancer cells. Following multiple steps of characterization and optimization, the anticancer activities of the LNPs were assessed against an aggressive breast cancer cell line, 4T1, in comparison with a normal cell line, LX-2. The selection of the helper phospholipid to be incorporated into the LNPs had a dramatic impact on their gene delivery performance. The optimized LNPs enabled a powerful MCT4 silencing by ∼90 % at low siRNA concentrations, with a subsequent ∼80 % cytotoxicity to 4T1 cells. Meanwhile, the LNPs demonstrated a 5-fold higher affinity to the breast cancer cells versus the normal cells, in which they had a minimum effect. Moreover, the MCT4 knockdown by the treatment remodeled the cytokine profile in 4T1 cells, as evidenced by 90 % and ∼64 % reduction in the levels of TNF-α and IL-6; respectively. The findings of this study are promising for potential clinical applications. Furthermore, the simple and scalable delivery vector developed herein can serve as a breast cancer-targeting platform for the delivery of other RNA therapeutics.
Collapse
Affiliation(s)
- Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Abdellatif Bouazzaoui
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Science and Technology Unit, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Department of Internal Medicine III (Haematology and Internal Oncology), University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
| | - Hesham M Tawfeek
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Mahmoud A Younis
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt.
| |
Collapse
|
4
|
Bai X, Smith ZL, Wang Y, Butterworth S, Tirella A. Sustained Drug Release from Smart Nanoparticles in Cancer Therapy: A Comprehensive Review. MICROMACHINES 2022; 13:mi13101623. [PMID: 36295976 PMCID: PMC9611581 DOI: 10.3390/mi13101623] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 05/14/2023]
Abstract
Although nanomedicine has been highly investigated for cancer treatment over the past decades, only a few nanomedicines are currently approved and in the market; making this field poorly represented in clinical applications. Key research gaps that require optimization to successfully translate the use of nanomedicines have been identified, but not addressed; among these, the lack of control of the release pattern of therapeutics is the most important. To solve these issues with currently used nanomedicines (e.g., burst release, systemic release), different strategies for the design and manufacturing of nanomedicines allowing for better control over the therapeutic release, are currently being investigated. The inclusion of stimuli-responsive properties and prolonged drug release have been identified as effective approaches to include in nanomedicine, and are discussed in this paper. Recently, smart sustained release nanoparticles have been successfully designed to safely and efficiently deliver therapeutics with different kinetic profiles, making them promising for many drug delivery applications and in specific for cancer treatment. In this review, the state-of-the-art of smart sustained release nanoparticles is discussed, focusing on the design strategies and performances of polymeric nanotechnologies. A complete list of nanomedicines currently tested in clinical trials and approved nanomedicines for cancer treatment is presented, critically discussing advantages and limitations with respect to the newly developed nanotechnologies and manufacturing methods. By the presented discussion and the highlight of nanomedicine design criteria and current limitations, this review paper could be of high interest to identify key features for the design of release-controlled nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Xue Bai
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Zara L. Smith
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Yuheng Wang
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Sam Butterworth
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Annalisa Tirella
- Division of Pharmacy and Optometry, School of Health Science, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- BIOtech-Center for Biomedical Technologies, Department of Industrial Engineering, University of Trento, Via delle Regole 101, 38123 Trento, Italy
- Correspondence:
| |
Collapse
|
5
|
Sargazi S, Arshad R, Ghamari R, Rahdar A, Bakhshi A, Karkan SF, Ajalli N, Bilal M, Díez-Pascual AM. siRNA-based nanotherapeutics as emerging modalities for immune-mediated diseases: A preliminary review. Cell Biol Int 2022; 46:1320-1344. [PMID: 35830711 PMCID: PMC9543380 DOI: 10.1002/cbin.11841] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 06/01/2022] [Accepted: 06/09/2022] [Indexed: 11/21/2022]
Abstract
Immune‐mediated diseases (IMDs) are chronic conditions that have an immune‐mediated etiology. Clinically, these diseases appear to be unrelated, but pathogenic pathways have been shown to connect them. While inflammation is a common occurrence in the body, it may either stimulate a favorable immune response to protect against harmful signals or cause illness by damaging cells and tissues. Nanomedicine has tremendous promise for regulating inflammation and treating IMIDs. Various nanoparticles coated with nanotherapeutics have been recently fabricated for effective targeted delivery to inflammatory tissues. RNA interference (RNAi) offers a tremendous genetic approach, particularly if traditional treatments are ineffective against IMDs. In cells, several signaling pathways can be suppressed by using RNAi, which blocks the expression of particular messenger RNAs. Using this molecular approach, the undesirable effects of anti‐inflammatory medications can be reduced. Still, there are many problems with using short‐interfering RNAs (siRNAs) to treat IMDs, including poor localization of the siRNAs in target tissues, unstable gene expression, and quick removal from the blood. Nanotherapeutics have been widely used in designing siRNA‐based carriers because of the restricted therapy options for IMIDs. In this review, we have discussed recent trends in the fabrication of siRNA nanodelivery systems, including lipid‐based siRNA nanocarriers, liposomes, and cationic lipids, stable nucleic acid‐lipid particles, polymeric‐based siRNA nanocarriers, polyethylenimine (PEI)‐based nanosystems, chitosan‐based nanoformulations, inorganic material‐based siRNA nanocarriers, and hybrid‐based delivery systems. We have also introduced novel siRNA‐based nanocarriers to control IMIDs, such as pulmonary inflammation, psoriasis, inflammatory bowel disease, ulcerative colitis, rheumatoid arthritis, etc. This study will pave the way for new avenues of research into the diagnosis and treatment of IMDs.
Collapse
Affiliation(s)
- Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Rabia Arshad
- Department of Pharmacy, Quaid-i-Azam University Islamabad, Islamabad, Pakistan
| | - Reza Ghamari
- Department of Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran
| | - Ali Bakhshi
- School of Physics, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Sonia Fathi Karkan
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Ajalli
- Department of Chemical Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, China
| | - Ana M Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Quimica Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
6
|
Kampel L, Goldsmith M, Ramishetti S, Veiga N, Rosenblum D, Gutkin A, Chatterjee S, Penn M, Lerman G, Peer D, Muhanna N. Therapeutic inhibitory RNA in head and neck cancer via functional targeted lipid nanoparticles. J Control Release 2021; 337:378-389. [PMID: 34303750 DOI: 10.1016/j.jconrel.2021.07.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 07/07/2021] [Accepted: 07/19/2021] [Indexed: 12/25/2022]
Abstract
Currently there are no specific therapies addressing the distinctive biology of human papillomavirus (HPV)-induced cancer approved for clinical use. Short interfering RNA (siRNA) has much potential for therapeutic manipulation of HPV E6/E7 oncoproteins. Lipid-based nanoparticles (LNPs) can be utilized for systemic transportation and delivery of siRNA at target site. We recently developed a recombinant protein linker that enables uniform conjugation of targeting antibodies to the LNPs. Herein, we demonstrate the therapeutic efficacy of anti-E6/E7 siRNA delivered via targeted LNPs (tLNPs) in a xenograft HPV-positive tumor model. We show that anti-epidermal growth factor receptor (EGFR) antibodies, anchored to the LNPs as targeting moieties, facilitate cargo delivery but also mediate anti-tumor activity. Treatment with siE6 via tLNPs resulted in 50% greater reduction of tumor volume compared to treatment with siControl encapsulated in isoLNPs (coated with isotype control antibodies). We demonstrate superior suppression of HPV oncogenes and higher induction of apoptosis by the tLNPs both in vitro and in vivo. Altogether, the coupling of inhibitory siE6 with anti-EGFR antibodies, that further elicited anti-tumor effects, successfully restricted tumor progression. This system that combines potent siRNA and therapeutically functional tLNPs can be modulated against various cancer models.
Collapse
Affiliation(s)
- Liyona Kampel
- The Head and Neck Cancer Research Laboratory, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel; The Department of Otolaryngology, Head and Neck Surgery and Maxillofacial Surgery, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel; Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Meir Goldsmith
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Srinivas Ramishetti
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nuphar Veiga
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniel Rosenblum
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Gutkin
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Sushmita Chatterjee
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Moran Penn
- The Head and Neck Cancer Research Laboratory, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel
| | - Galya Lerman
- The Head and Neck Cancer Research Laboratory, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Nidal Muhanna
- The Head and Neck Cancer Research Laboratory, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel; The Department of Otolaryngology, Head and Neck Surgery and Maxillofacial Surgery, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel.
| |
Collapse
|
7
|
Badu S, Melnik R, Singh S. Mathematical and computational models of RNA nanoclusters and their applications in data-driven environments. MOLECULAR SIMULATION 2020. [DOI: 10.1080/08927022.2020.1804564] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Shyam Badu
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, Waterloo, Ontario, Canada
| | - Roderick Melnik
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, Waterloo, Ontario, Canada
- BCAM-Basque Center for Applied Mathematics, Bilbao, Spain
| | - Sundeep Singh
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, Waterloo, Ontario, Canada
| |
Collapse
|
8
|
Paris JL, Coelho F, Teixeira A, Diéguez L, Silva BFB, Abalde-Cela S. In Vitro Evaluation of Lipopolyplexes for Gene Transfection: Comparing 2D, 3D and Microdroplet-Enabled Cell Culture. Molecules 2020; 25:molecules25143277. [PMID: 32708478 PMCID: PMC7397275 DOI: 10.3390/molecules25143277] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/06/2020] [Accepted: 07/14/2020] [Indexed: 01/11/2023] Open
Abstract
Complexes combining nucleic acids with lipids and polymers (lipopolyplexes) show great promise for gene therapy since they enable compositional, physical and functional versatility to be optimized for therapeutic efficiency. When developing lipopolyplexes for gene delivery, one of the first evaluations performed is an in vitro transfection efficiency experiment. Many different in vitro models can be used, and the effect of the model on the experiment outcome has not been thoroughly studied. The objective of this work was to compare the insights obtained from three different in vitro models, as well as the potential limitations associated with each of them. We have prepared a series of lipopolyplex formulations with three different cationic polymers (poly-l-lysine, bioreducible poly-l-lysine and polyethyleneimine), and assessed their in vitro biological performance in 2D monolayer cell culture, 3D spheroid culture and microdroplet-based single-cell culture. Lipopolyplexes from different polymers presented varying degrees of transfection efficiency in all models. The best-performing formulation in 2D culture was the polyethyleneimine lipopolyplex, while lipoplexes prepared with bioreducible poly-l-lysine were the only ones achieving any transfection in microdroplet-enabled cell culture. None of the prepared formulations achieved significant gene transfection in 3D culture. All of the prepared formulations were well tolerated by cells in 2D culture, while at least one formulation (poly-l-lysine polyplex) delayed 3D spheroid growth. These results highlight the need for selecting the appropriate in vitro model depending on the intended application.
Collapse
|
9
|
Romano S, Fonseca N, Simões S, Gonçalves J, Moreira JN. Nucleolin-based targeting strategies for cancer therapy: from targeted drug delivery to cytotoxic ligands. Drug Discov Today 2019; 24:1985-2001. [PMID: 31271738 DOI: 10.1016/j.drudis.2019.06.018] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 06/08/2019] [Accepted: 06/26/2019] [Indexed: 01/10/2023]
Abstract
Cancer is currently the second leading cause of death worldwide and current therapeutic approaches remain ineffective in several cases. Therefore, there is a need to develop more efficacious therapeutic agents, especially for subtypes of cancer lacking targeted therapies. Limited drug penetration into tumors impairs the efficacy of therapies targeting cancer cells. One of the strategies to overcome this problem is targeting the more accessible tumor vasculature via molecules such as nucleolin, which is expressed at the surface of cancer and angiogenic endothelial cells, thus enabling a dual cellular targeting strategy. In this review, we present and discuss nucleolin-based targeting strategies that have been developed for cancer therapy, with a special focus on recent antibody-based approaches.
Collapse
Affiliation(s)
- Sofia Romano
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine (Pólo I), University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Nuno Fonseca
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine (Pólo I), University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; TREAT U, SA, Parque Industrial de Taveiro, Lote 44, 3045-508 Coimbra, Portugal
| | - Sérgio Simões
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine (Pólo I), University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548 Portugal
| | - João Gonçalves
- iMed. ULisboa - Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Avenida Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - João Nuno Moreira
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine (Pólo I), University of Coimbra, Rua Larga, 3004-504, Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548 Portugal.
| |
Collapse
|
10
|
Kanehira Y, Togami K, Ishizawa K, Sato S, Tada H, Chono S. Intratumoral delivery and therapeutic efficacy of nanoparticle-encapsulated anti-tumor siRNA following intrapulmonary administration for potential treatment of lung cancer. Pharm Dev Technol 2019; 24:1095-1103. [DOI: 10.1080/10837450.2019.1633345] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yukimune Kanehira
- Division of Pharmaceutics, Hokkaido Pharmaceutical University School of Pharmacy, Sapporo, Japan
| | - Kohei Togami
- Division of Pharmaceutics, Hokkaido Pharmaceutical University School of Pharmacy, Sapporo, Japan
- Faculty of Pharmaceutical Sciences, Department of Pharmaceutics, Hokkaido University of Science, Sapporo, Japan
| | - Kiyomi Ishizawa
- Division of Pharmaceutics, Hokkaido Pharmaceutical University School of Pharmacy, Sapporo, Japan
- Faculty of Pharmaceutical Sciences, Department of Pharmaceutics, Hokkaido University of Science, Sapporo, Japan
| | - Shingo Sato
- Division of Pharmaceutics, Hokkaido Pharmaceutical University School of Pharmacy, Sapporo, Japan
| | - Hitoshi Tada
- Division of Pharmaceutics, Hokkaido Pharmaceutical University School of Pharmacy, Sapporo, Japan
- Faculty of Pharmaceutical Sciences, Department of Pharmaceutics, Hokkaido University of Science, Sapporo, Japan
| | - Sumio Chono
- Division of Pharmaceutics, Hokkaido Pharmaceutical University School of Pharmacy, Sapporo, Japan
- Faculty of Pharmaceutical Sciences, Department of Pharmaceutics, Hokkaido University of Science, Sapporo, Japan
| |
Collapse
|
11
|
Tieu T, Alba M, Elnathan R, Cifuentes‐Rius A, Voelcker NH. Advances in Porous Silicon–Based Nanomaterials for Diagnostic and Therapeutic Applications. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800095] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Terence Tieu
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Maria Alba
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| | - Roey Elnathan
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| | - Anna Cifuentes‐Rius
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Campus, 381 Royal Parade Parkville Victoria 3052 Australia
- Prof. N. H. Voelcker Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility 151 Wellington Road Clayton Victoria 3168 Australia
- T. Tieu, Dr. M. Alba, Prof. N. H. Voelcker CSIRO Manufacturing Bayview Avenue Clayton Victoria 3168 Australia
| |
Collapse
|
12
|
|
13
|
Sakurai Y, Hada T, Harashima H. Scalable preparation of poly(ethylene glycol)-grafted siRNA-loaded lipid nanoparticles using a commercially available fluidic device and tangential flow filtration. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:1086-1096. [PMID: 28157422 DOI: 10.1080/09205063.2017.1291297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
While a number of siRNA delivery systems have been developed, the methods used in their preparation are not suitable for large-scale production. We herein report on methodology for the large-scale preparation of liposomal siRNA using a fluidic device and tangential flow filtration (TFF). A number of studies have appeared on the use of fluidic devices for preparing and purifying liposomes, but no systematic information regarding appropriate membrane type of commercially available apparatus is available. The findings reported herein indicate that, under optimized conditions, a microfluidic device and TFF can be used to produce siRNA lipid nanoparticles with the same characteristics as traditional ones'. The in vivo silencing efficiency of these lipid nanoparticles in the liver was comparable to laboratory-produced nanoparticles. In addition, con-focal laser scanning microscopy analyses revealed that they accumulated in the liver accumulation at the same levels as particles produced by batch-type and continuous-type procedures. This methodology has the potential to contribute to the advancement of this process from basic research to clinical studies of liposomal DDS.
Collapse
Affiliation(s)
- Yu Sakurai
- a Faculty of Pharmaceutical Sciences , Hokkaido University , Sapporo , Japan
| | - Tomoya Hada
- a Faculty of Pharmaceutical Sciences , Hokkaido University , Sapporo , Japan
| | - Hideyoshi Harashima
- a Faculty of Pharmaceutical Sciences , Hokkaido University , Sapporo , Japan
| |
Collapse
|
14
|
Ramot Y, Rotkopf S, Gabai RM, Zorde Khvalevsky E, Muravnik S, Marzoli GA, Domb AJ, Shemi A, Nyska A. Preclinical Safety Evaluation in Rats of a Polymeric Matrix Containing an siRNA Drug Used as a Local and Prolonged Delivery System for Pancreatic Cancer Therapy. Toxicol Pathol 2016; 44:856-65. [PMID: 27147553 DOI: 10.1177/0192623316645860] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Conventional chemotherapy treatments for pancreatic cancer are mainly palliative. RNA interference (RNAi)-based drugs present the potential for a new targeted treatment. LOcal Drug EluteR (LODER(TM)) is a novel biodegradable polymeric matrix that shields drugs against enzymatic degradation and releases small interfering RNA (siRNA) against G12D-mutated KRAS (siG12D). siG12D-LODER has successfully passed a phase 1/2a clinical trial. Such a formulation necessitates biocompatibility and safety studies. We describe the safety and toxicity studies with siG12D-LODER in 192 Hsd:Sprague Dawley rats, after repeated subcutaneous administrations (days 1, 14, and 28). Animals were sacrificed on days 29 and 42 (recovery phase). In all groups, no adverse effects were noted, and all animals showed favorable local and systemic tolerability. Histopathologically, LODER implantation resulted in the expected capsule formation, composed of a thin fibrotic tissue. On the interface between the cavity and the capsule, a single layer composed of macrophages and multinucleated giant cells was observed. No difference was noted between the placebo and siG12D-LODER groups. These findings provide valuable information for future preclinical studies with siRNA-bearing biodegradable polymers and for the safety aspects of RNAi-based drugs as a targeted therapy.
Collapse
Affiliation(s)
- Yuval Ramot
- Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | - Abraham J Domb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, Jerusalem, Israel
| | | | - Abraham Nyska
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel Consultant in Toxicologic Pathology, Timrat, Israel
| |
Collapse
|
15
|
Ngamcherdtrakul W, Castro DJ, Gu S, Morry J, Reda M, Gray JW, Yantasee W. Current development of targeted oligonucleotide-based cancer therapies: Perspective on HER2-positive breast cancer treatment. Cancer Treat Rev 2016; 45:19-29. [PMID: 26930249 DOI: 10.1016/j.ctrv.2016.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/13/2016] [Accepted: 02/15/2016] [Indexed: 12/24/2022]
Abstract
This Review discusses the various types of non-coding oligonucleotides, which have garnered extensive interest as new alternatives for targeted cancer therapies over small molecule inhibitors and monoclonal antibodies. These oligonucleotides can target any hallmark of cancer, no longer limited to so-called "druggable" targets. Thus, any identified gene that plays a key role in cancer progression or drug resistance can be exploited with oligonucleotides. Among them, small-interfering RNAs (siRNAs) are frequently utilized for gene silencing due to the robust and well established mechanism of RNA interference. Despite promising advantages, clinical translation of siRNAs is hindered by the lack of effective delivery platforms. This Review provides general criteria and consideration of nanoparticle development for systemic siRNA delivery. Different classes of nanoparticle candidates for siRNA delivery are discussed, and the progress in clinical trials for systemic cancer treatment is reviewed. Lastly, this Review presents HER2 (human epidermal growth factor receptor type 2)-positive breast cancer as one example that could benefit significantly from siRNA technology. How siRNA-based therapeutics can overcome cancer resistance to such therapies is discussed.
Collapse
Affiliation(s)
- Worapol Ngamcherdtrakul
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA; PDX Pharmaceuticals, LLC, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - David J Castro
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA; PDX Pharmaceuticals, LLC, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Shenda Gu
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Jingga Morry
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Moataz Reda
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Joe W Gray
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA.
| | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA; PDX Pharmaceuticals, LLC, 3303 SW Bond Ave, Portland, OR 97239, USA.
| |
Collapse
|
16
|
Zuckerman JE, Davis ME. Clinical experiences with systemically administered siRNA-based therapeutics in cancer. Nat Rev Drug Discov 2015; 14:843-56. [PMID: 26567702 DOI: 10.1038/nrd4685] [Citation(s) in RCA: 318] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Small interfering RNA (siRNA)-based therapies are emerging as a promising new anticancer approach, and a small number of Phase I clinical trials involving patients with solid tumours have now been completed. Encouraging results from these pioneering clinical studies show that these new therapeutics can successfully and safely inhibit targeted gene products in patients with cancer, and have taught us important lessons regarding appropriate dosages and schedules. In this Review, we critically assess these Phase I studies and discuss their implications for future clinical trial design. Key challenges and future directions in the development of siRNA-containing anticancer therapeutics are also considered.
Collapse
Affiliation(s)
- Jonathan E Zuckerman
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Mark E Davis
- Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
17
|
Young SWS, Stenzel M, Yang JL. Nanoparticle-siRNA: A potential cancer therapy? Crit Rev Oncol Hematol 2015; 98:159-69. [PMID: 26597018 DOI: 10.1016/j.critrevonc.2015.10.015] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/27/2015] [Indexed: 01/04/2023] Open
Abstract
PURPOSE To explore current developments in short interfering RNA (siRNA) delivery systems in nanooncology, in particular nanoparticles that encapsulate siRNA for targeted treatment of cancer. siRNA has a high specificity towards the oncogenic mRNA in cancer cells, while application of nanoparticles can improve stable delivery and enhance efficacy. METHODS A literature search was performed using the terms "siRNA", "nanoparticles", "targeted delivery", and "cancer". These databases included Medline, Embase, Cochrane Review, Pubmed, and Scopus. RESULTS siRNA anti-cancer drugs utilize endogenous RNAi mechanisms to silence oncogene expression, which promotes cancer remission. However, current delivery methods have poor efficacy, requiring assistance by nanoparticles for successful delivery. Recently several preclinical studies have crossed into clinical trials utilizing siRNA nanoparticle therapeutics. CONCLUSION Great potential exists for nano-siRNA drugs in cancer treatment, but issues exist with nanoparticle toxicity and off target siRNA effects. Further research is needed in this rapidly developing and promising field of nano-siRNA drugs.
Collapse
Affiliation(s)
- Samuel Wang Sherng Young
- Adult Cancer Program, Lowy Cancer Research Centre, Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Martina Stenzel
- Centre for Advanced Macromolecular Design, Faculty of Science, University of New South Wales, Sydney, NSW, Australia
| | - Jia-Lin Yang
- Adult Cancer Program, Lowy Cancer Research Centre, Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
18
|
Effective Small Interfering RNA Therapy to Treat CLCN7-dependent Autosomal Dominant Osteopetrosis Type 2. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e248. [PMID: 26325626 PMCID: PMC4877447 DOI: 10.1038/mtna.2015.21] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/15/2015] [Indexed: 01/14/2023]
Abstract
In about 70% of patients affected by autosomal dominant osteopetrosis type 2 (ADO2), osteoclast activity is reduced by heterozygous mutations of the CLCN7 gene, encoding the ClC-7 chloride/hydrogen antiporter. CLCN7(G215R)-, CLCN7(R767W)-, and CLCN7(R286W)-specific siRNAs silenced transfected mutant mRNA/EGFP in HEK293 cells, in RAW264.7 cells and in human osteoclasts, with no change of CLCN7(WT) mRNA and no effect of scrambled siRNA on the mutant transcripts. Osteoclasts from Clcn7(G213R) ADO2 mice showed reduced bone resorption, a condition rescued by Clcn7(G213R)-specific siRNA. Treatment of ADO2 mice with Clcn7(G213R)-specific siRNA induced increase of bone resorption variables and decrease of trabecular bone mass, leading to an overall improvement of the osteopetrotic bone phenotype. Treatment did not induce overt adverse effects and was effective also with siRNAs specific for other mutants. These results demonstrate that a siRNA-based experimental treatment of ADO2 is feasible, and underscore a translational impact for future strategy to cure this therapeutically neglected form of osteopetrosis.
Collapse
|
19
|
Abstract
Nanoparticle (NP) delivery systems for small interfering RNA (siRNA) that have good systemic circulation and high nucleic acid content are highly desired for translation into clinical use. Here, a family of cationic mucic acid-containing polymers is synthesized and shown to assemble with siRNA to form NPs. A cationic mucic acid polymer (cMAP) containing alternating mucic acid and charged monomers is synthesized. When combined with siRNA, cMAP forms NPs that require steric stabilization by poly(ethylene glycol) (PEG) that is attached to the NP surface via a 5-nitrophenylboronic acid linkage (5-nitrophenylboronic acid-PEGm (5-nPBA-PEGm)) to diols on mucic acid in the cMAP in order to inhibit aggregation in biological fluids. As an alternative, cMAP is covalently conjugated with PEG via two methods. First, a copolymer is prepared with alternating cMAP-PEG units that can form loops of PEG on the surface of the formulated siRNA-containing NPs. Second, an mPEG-cMAP-PEGm triblock polymer is synthesized that could lead to a PEG brush configuration on the surface of the formulated siRNA-containing NPs. The copolymer and triblock polymer are able to form stable siRNA-containing NPs without and with the addition of 5-nPBA-PEGm. Five formulations, (i) cMAP with 5-nPBA-PEGm, (ii) cMAP-PEG copolymer both (a) with and (b) without 5-nPBA-PEGm, and (iii) mPEG-cMAP-PEGm triblock polymer both (a) with and (b) without 5-nPBA-PEGm, are used to produce NPs in the 30-40 nm size range, and their circulation times are evaluated in mice using tail vein injections. The mPEG-cMAP-PEGm triblock polymer provides the siRNA-containing NP with the longest circulation time (5-10% of the formulation remains in circulation at 60 min postdosing), even when a portion of the excess cationic components used in the formulation is filtered away prior to injection. A NP formulation using the mPEG-cMAP-PEGm triblock polymer that is free of excess components could contain as much as ca. 30 wt % siRNA.
Collapse
Affiliation(s)
- Dorothy W Pan
- Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Mark E Davis
- Chemical Engineering, California Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
20
|
Wang HC, Yang Y, Xu SY, Peng J, Jiang JH, Li CY. The CRISPR/Cas system inhibited the pro-oncogenic effects of alternatively spliced fibronectin extra domain A via editing the genome in salivary adenoid cystic carcinoma cells. Oral Dis 2015; 21:608-18. [PMID: 25684411 DOI: 10.1111/odi.12323] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/24/2015] [Accepted: 02/02/2015] [Indexed: 12/17/2022]
Affiliation(s)
- H-C Wang
- The Central Laboratory; Peking University School and Hospital of Stomatology; Haidian District Beijing China
| | - Y Yang
- The Central Laboratory; Peking University School and Hospital of Stomatology; Haidian District Beijing China
| | - S-Y Xu
- Department of Oral Implanting; Shandong University School of Stomatology; Lixia District Jinan China
| | - J Peng
- The Central Laboratory; Peking University School and Hospital of Stomatology; Haidian District Beijing China
| | - J-H Jiang
- The Department of Orthodontics; Peking University School and Hospital of Stomatology; Haidian District China
| | - C-Y Li
- The Central Laboratory; Peking University School and Hospital of Stomatology; Haidian District Beijing China
| |
Collapse
|
21
|
Asanuma H, Kashida H, Kamiya Y. De novo design of functional oligonucleotides with acyclic scaffolds. CHEM REC 2014; 14:1055-69. [PMID: 25171046 DOI: 10.1002/tcr.201402040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Indexed: 01/20/2023]
Abstract
In this account, we demonstrate a new methodology for the de novo design of functional oligonucleotides with the acyclic scaffolds threoninol and serinol. Four functional motifs-wedge, interstrand-wedge, dimer, and cluster-have been prepared from natural DNA or RNA and functional base surrogates prepared from d-threoninol. The following applications of these motifs are described: (1) photoregulation of formation and dissociation of a DNA duplex modified with azobenzene, (2) sequence-specific detection of DNA using a fluorescent probe, (3) formation of fluorophore assemblies that mimic quantum dots, (4) improved strand selectivity of siRNA modified with a base surrogate, and (5) in vivo tracing of the RNAi pathway. Finally, we introduce artificial nucleic acids (XNAs) prepared from d-threoninol and serinol functionalized with each of the four nucleobases, which have unique properties compared with other acyclic XNAs. Functional oligonucleotides designed from acyclic scaffolds will be powerful tools for both DNA nanotechnology and biotechnology.
Collapse
Affiliation(s)
- Hiroyuki Asanuma
- Department of Molecular Design and Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan.
| | | | | |
Collapse
|