1
|
Beigi A, Naghib SM, Matini A, Tajabadi M, Mozafari MR. Lipid-Based Nanocarriers for Targeted Gene Delivery in Lung Cancer Therapy: Exploring a Novel Therapeutic Paradigm. Curr Gene Ther 2025; 25:92-112. [PMID: 38778601 DOI: 10.2174/0115665232292768240503050508] [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: 11/14/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 05/25/2024]
Abstract
Lung cancer is a significant cause of cancer-related death worldwide. It can be broadly categorised into small-cell lung cancer (SCLC) and Non-small cell lung cancer (NSCLC). Surgical intervention, radiation therapy, and the administration of chemotherapeutic medications are among the current treatment modalities. However, the application of chemotherapy may be limited in more advanced stages of metastasis due to the potential for adverse effects and a lack of cell selectivity. Although small-molecule anticancer treatments have demonstrated effectiveness, they still face several challenges. The challenges at hand in this context comprise insufficient solubility in water, limited bioavailability at specific sites, adverse effects, and the requirement for epidermal growth factor receptor inhibitors that are genetically tailored. Bio-macromolecular drugs, including small interfering RNA (siRNA) and messenger RNA (mRNA), are susceptible to degradation when exposed to the bodily fluids of humans, which can reduce stability and concentration. In this context, nanoscale delivery technologies are utilised. These agents offer encouraging prospects for the preservation and regulation of pharmaceutical substances, in addition to improving the solubility and stability of medications. Nanocarrier-based systems possess the notable advantage of facilitating accurate and sustained drug release, as opposed to traditional systemic methodologies. The primary focus of scientific investigation has been to augment the therapeutic efficacy of nanoparticles composed of lipids. Numerous nanoscale drug delivery techniques have been implemented to treat various respiratory ailments, such as lung cancer. These technologies have exhibited the potential to mitigate the limitations associated with conventional therapy. As an illustration, applying nanocarriers may enhance the solubility of small-molecule anticancer drugs and prevent the degradation of bio-macromolecular drugs. Furthermore, these devices can administer medications in a controlled and extended fashion, thereby augmenting the therapeutic intervention's effectiveness and reducing adverse reactions. However, despite these promising results, challenges remain that must be addressed. Multiple factors necessitate consideration when contemplating the application of nanoparticles in medical interventions. To begin with, the advancement of more efficient delivery methods is imperative. In addition, a comprehensive investigation into the potential toxicity of nanoparticles is required. Finally, additional research is needed to comprehend these treatments' enduring ramifications. Despite these challenges, the field of nanomedicine demonstrates considerable promise in enhancing the therapy of lung cancer and other respiratory diseases.
Collapse
Affiliation(s)
- Anahita Beigi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Amir Matini
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Maryam Tajabadi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran, 16844, Iran
| | - Mohammad Reza Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
2
|
Liu J, Han H, Yang B, Zhang N, Li J, Chen X, Wu J, Zhao Y, Yang Y. Immunogenicity and protective efficacy of the HC009 mRNA vaccine against SARS-CoV-2. Front Immunol 2024; 15:1416375. [PMID: 39131158 PMCID: PMC11310568 DOI: 10.3389/fimmu.2024.1416375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/27/2024] [Indexed: 08/13/2024] Open
Abstract
With the rapid global spread of COVID-19 and the continuous emergence of variants, there is an urgent need to develop safe and effective vaccines. Here, we developed a novel mRNA vaccine, HC009, based on new formulation by the QTsome delivery platform. Immunogenicity results showed that the prime-boost immunization strategy with HC009 was able to induce robust and durable humoral immunity, as well as Th1-biased cellular responses in rodents or non-human primates (NHPs). After further challenge with live SARS-CoV-2 virus, HC009 provided adequate protection against virus infection in hACE2 transgenic mice. Therefore, HC009 could provide significant immune protection against SARS-CoV-2.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- COVID-19 Vaccines/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Mice
- mRNA Vaccines/immunology
- Mice, Transgenic
- Immunogenicity, Vaccine
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Humans
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Immunity, Humoral
- Female
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Mice, Inbred BALB C
- Vaccine Efficacy
Collapse
Affiliation(s)
- Juan Liu
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | | - Yongsheng Yang
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Omidian H, Gill EJ, Cubeddu LX. Lipid Nanoparticles in Lung Cancer Therapy. Pharmaceutics 2024; 16:644. [PMID: 38794306 PMCID: PMC11124812 DOI: 10.3390/pharmaceutics16050644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/22/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
This manuscript explores the use of lipid nanoparticles (LNPs) in addressing the pivotal challenges of lung cancer treatment, including drug delivery inefficacy and multi-drug resistance. LNPs have significantly advanced targeted therapy by improving the precision and reducing the systemic toxicity of chemotherapeutics such as doxorubicin and paclitaxel. This manuscript details the design and benefits of various LNP systems, including solid lipid-polymer hybrids, which offer controlled release and enhanced drug encapsulation. Despite achievements in reducing tumor size and enhancing survival, challenges such as manufacturing complexity, biocompatibility, and variable clinical outcomes persist. Future directions are aimed at refining targeting capabilities, expanding combinatorial therapies, and integrating advanced manufacturing techniques to tailor treatments to individual patient profiles, thus promising to transform lung cancer therapy through interdisciplinary collaboration and regulatory innovation.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | | | - Luigi X. Cubeddu
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| |
Collapse
|
4
|
Wang R, Yi L, Zhou W, Wang W, Wang L, Xu L, Deng C, He M, Xie Y, Xu J, Chen Y, Gao T, Jin Q, Zhang L, Xie M. Targeted microRNA delivery by lipid nanoparticles and gas vesicle-assisted ultrasound cavitation to treat heart transplant rejection. Biomater Sci 2023; 11:6492-6503. [PMID: 36884313 DOI: 10.1039/d2bm02103j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Despite exquisite immune response modulation, the extensive application of microRNA therapy in treating heart transplant rejection is still impeded by poor stability and low target efficiency. Here we have developed a low-intensity pulsed ultrasound (LIPUS) cavitation-assisted genetic therapy after executing the heart transplantation (LIGHT) strategy, facilitating microRNA delivery to target tissues through the LIPUS cavitation of gas vesicles (GVs), a class of air-filled protein nanostructures. We prepared antagomir-155 encapsulated liposome nanoparticles to enhance the stability. Then the murine heterotopic transplantation model was established, and antagomir-155 was delivered to murine allografted hearts via the cavitation of GVs agitated by LIPUS, which reinforced the target efficiency while guaranteeing safety owing to the specific acoustic property of GVs. This LIGHT strategy significantly depleted miR-155, upregulating the suppressors of cytokine signaling 1 (SOCS1), leading to reparative polarization of macrophages, decrease of T lymphocytes and reduction of inflammatory factors. Thereby, rejection was attenuated and the allografted heart survival was markedly prolonged. The LIGHT strategy achieves targeted delivery of microRNA with minimal invasiveness and great efficiency, paving the way towards novel ultrasound cavitation-assisted strategies of targeted genetic therapy for heart transplantation rejection.
Collapse
Affiliation(s)
- Rui Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Luyang Yi
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Wuqi Zhou
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Wenyuan Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Lufang Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Lingling Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Mengrong He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Yuji Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Jia Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Tang Gao
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| |
Collapse
|
5
|
Abdellatif AAH, Scagnetti G, Younis MA, Bouazzaoui A, Tawfeek HM, Aldosari BN, Almurshedi AS, Alsharidah M, Rugaie OA, Davies MPA, Liloglou T, Ross K, Saleem I. Non-coding RNA-directed therapeutics in lung cancer: Delivery technologies and clinical applications. Colloids Surf B Biointerfaces 2023; 229:113466. [PMID: 37515959 DOI: 10.1016/j.colsurfb.2023.113466] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/28/2023] [Accepted: 07/16/2023] [Indexed: 07/31/2023]
Abstract
Lung cancer is one of the most aggressive and deadliest health threats. There has been an increasing interest in non-coding RNA (ncRNA) recently, especially in the areas of carcinogenesis and tumour progression. However, ncRNA-directed therapies are still encountering obstacles on their way to the clinic. In the present article, we provide an overview on the potential of targeting ncRNA in the treatment of lung cancer. Then, we discuss the delivery challenges and recent approaches enabling the delivery of ncRNA-directed therapies to the lung cancer cells, where we illuminate some advanced technologies including chemically-modified oligonucleotides, nuclear targeting, and three-dimensional in vitro models. Furthermore, advanced non-viral delivery systems recruiting nanoparticles, biomimetic delivery systems, and extracellular vesicles are also highlighted. Lastly, the challenges limiting the clinical trials on the therapeutic targeting of ncRNAs in lung cancer and future directions to tackle them are explored.
Collapse
Affiliation(s)
- Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Al Qassim 51452, Saudi Arabia; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Giulia Scagnetti
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Liverpool L3 3AF, UK
| | - Mahmoud A Younis
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, 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; Medical Clinic, Hematology/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
| | - Basmah N Aldosari
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alanood S Almurshedi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mansour Alsharidah
- Department of Physiology, College of Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| | - Osamah Al Rugaie
- Department of Basic Medical Sciences, College of Medicine and Medical Sciences, Qassim University, P.O. Box 991, Unaizah, Al Qassim 51911, Saudi Arabia
| | - Michael P A Davies
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular & Integrative Biology, The University of Liverpool, UK
| | | | - Kehinde Ross
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Liverpool L3 3AF, UK; Institute for Health Research, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - Imran Saleem
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, James Parsons Building, Liverpool L3 3AF, UK; Institute for Health Research, Liverpool John Moores University, Liverpool L3 3AF, UK.
| |
Collapse
|
6
|
Lee DY, Amirthalingam S, Lee C, Rajendran AK, Ahn YH, Hwang NS. Strategies for targeted gene delivery using lipid nanoparticles and cell-derived nanovesicles. NANOSCALE ADVANCES 2023; 5:3834-3856. [PMID: 37496613 PMCID: PMC10368001 DOI: 10.1039/d3na00198a] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/10/2023] [Indexed: 07/28/2023]
Abstract
Gene therapy is a promising approach for the treatment of many diseases. However, the effective delivery of the cargo without degradation in vivo is one of the major hurdles. With the advent of lipid nanoparticles (LNPs) and cell-derived nanovesicles (CDNs), gene delivery holds a very promising future. The targeting of these nanosystems is a prerequisite for effective transfection with minimal side-effects. In this review, we highlight the emerging strategies utilized for the effective targeting of LNPs and CDNs, and we summarize the preparation methodologies for LNPs and CDNs. We have also highlighted the non-ligand targeting of LNPs toward certain organs based on their composition. It is highly expected that continuing the developments in the targeting approaches of LNPs and CDNs for the delivery system will further promote them in clinical translation.
Collapse
Affiliation(s)
- Dong-Yup Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
- Institute of Engineering Research, Seoul National University Seoul 08826 Republic of Korea
| | - Changyub Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Arun Kumar Rajendran
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
| | - Young-Hyun Ahn
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University Seoul 08826 Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University Seoul 08826 Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University Seoul 08826 Republic of Korea
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University Seoul 08826 Republic of Korea
- Institute of Engineering Research, Seoul National University Seoul 08826 Republic of Korea
| |
Collapse
|
7
|
Min SH, Lei W, Jun CJ, Yan ZS, Guang YX, Tong Z, Yong ZP, Hui LZ, Xing H. Design strategy and research progress of multifunctional nanoparticles in lung cancer therapy. Expert Opin Investig Drugs 2023; 32:723-739. [PMID: 37668152 DOI: 10.1080/13543784.2023.2254683] [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: 04/25/2023] [Revised: 08/01/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
INTRODUCTION Lung cancer is one of the cancer types with the highest mortality rate, exploring a more effective treatment modality that improves therapeutic efficacy while mitigating side effects is now an urgent requirement. Designing multifunctional nanoparticles can be used to overcome the limitations of drugs and conventional drug delivery systems. Nanotechnology has been widely researched, and through different needs, suitable nanocarriers can be selected to load anti-cancer drugs to improve the therapeutic effect. It is foreseeable that with the rapid development of nanotechnology, more and more lung cancer patients will benefit from nanotechnology. This paper reviews the merits of various multifunctional nanoparticles in the treatment of lung cancer to provide novel ideas for lung cancer treatment. AREAS COVERED This review focuses on summarizing various nanoparticles for targeted lung cancer therapy and their advantages and disadvantages, using nanoparticles loaded with anti-cancer drugs, delivered to lung cancer sites, enhancing drug half-life, improving anti-cancer drug efficacy and reducing side effects. EXPERT OPINION The delivery mode of nanoparticles with superior pharmacokinetic properties in the in vivo circulation enhances the half-life of the drug, and provides tissue-targeted selectivity and the ability to overcome biological barriers, bringing a revolution in the field of oncology.
Collapse
Affiliation(s)
- Shen Hui Min
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Lei
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Jia Jun
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhang Shao Yan
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Xu Guang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhang Tong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheng Pei Yong
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Zhen Hui
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huang Xing
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
8
|
Subramaniam S, Joyce P, Donnellan L, Young C, Wignall A, Hoffmann P, Prestidge CA. Protein adsorption determines pulmonary cell uptake of lipid-based nanoparticles. J Colloid Interface Sci 2023; 641:36-47. [PMID: 36924544 DOI: 10.1016/j.jcis.2023.03.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
The inhalable administration of lipid nanoparticles is an effective strategy for localised delivery of therapeutics against various lung diseases. Of this, improved intracellular delivery of pharmaceuticals for infectious disease and cancer management is of high significance. However, the influence of lipid nanoparticle composition and structure on uptake in pulmonary cell lines, especially in the presence of biologically relevant media is poorly understood. Here, the uptake of lamellar (liposomes) versus non-lamellar (cubosomes) lipid nanoparticles in macrophages and lung epithelial cells was quantified and the influence of bronchoalveolar lavage fluid (BALF), containing native pulmonary protein and surfactant molecules is determined. Cubosome uptake in both macrophages and epithelial cells was strongly mediated by a high percentage of molecular function regulatory and binding proteins present within the protein corona. In contrast, the protein corona did not influence the uptake of liposomes in epithelial cells. In macrophages, the proteins mediated a rapid internalisation, followed by exocytosis of liposomes after 6 h incubation. These findings on the influence of biological fluid in regulating lipid nanoparticle uptake mechanisms may guide future development of optimal intracellular delivery systems for therapeutics via the pulmonary route.
Collapse
Affiliation(s)
- Santhni Subramaniam
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Paul Joyce
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Leigh Donnellan
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Clifford Young
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Anthony Wignall
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Peter Hoffmann
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Clive A Prestidge
- University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia.
| |
Collapse
|
9
|
Murugan D, Rangasamy L. A perspective to weaponize microRNAs against lung cancer. Noncoding RNA Res 2023; 8:18-32. [PMID: 36262424 PMCID: PMC9556932 DOI: 10.1016/j.ncrna.2022.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 12/02/2022] Open
Abstract
microRNAs are regulatory RNAs that silence specific mRNA by binding to it, inducing translational repression. Over the recent decades since the discovery of RNA interference, the field of microRNA therapeutics has expanded tremendously. The role of miRNAs in disease development has attracted researchers to investigate their potential in therapeutics. In lung cancer, multiple miRNAs are deregulated, and their involvement is observed in cell proliferation, immunomodulation, angiogenesis, and epithelial-mesenchymal transition. Thus, synthetic oligonucleotides are developed to downregulate the overexpressed miRNA or to upregulate the repressed miRNA. However, their clinical efficiency is limited due to the requirement for an effective delivery strategy. Advances in the current understanding of nanotechnology, biomaterial science, and disease molecular pathology have increased the chances of overcoming the limitations of miRNA-based therapy. This review enlists downregulated and upregulated miRNAs in lung cancer. This review also highlights the major contributions to miRNA-based therapeutics for lung cancer and strategies to overcome endosomal barriers. It also attempts to understand the nuances between current advancements in delivery methods, advantages, disadvantages, and practical issues for the large-scale development of miRNA-based therapeutics.
Collapse
Affiliation(s)
- Dhanashree Murugan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular, and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
- School of Biosciences & Technology (SBST), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| | - Loganathan Rangasamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular, and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, 632014, Tamil Nadu, India
| |
Collapse
|
10
|
Gaobotse G, Venkataraman S, Brown PD, Masisi K, Kwape TE, Nkwe DO, Rantong G, Makhzoum A. The use of African medicinal plants in cancer management. Front Pharmacol 2023; 14:1122388. [PMID: 36865913 PMCID: PMC9971233 DOI: 10.3389/fphar.2023.1122388] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Cancer is the third leading cause of premature death in sub-Saharan Africa. Cervical cancer has the highest number of incidences in sub-Saharan Africa due to high HIV prevalence (70% of global cases) in African countries which is linked to increasing the risk of developing cervical cancer, and the continuous high risk of being infected with Human papillomavirus In 2020, the risk of dying from cancer amongst women was higher in Eastern Africa (11%) than it was in Northern America (7.4%). Plants continue to provide unlimited pharmacological bioactive compounds that are used to manage various illnesses, including cancer. By reviewing the literature, we provide an inventory of African plants with reported anticancer activity and evidence supporting their use in cancer management. In this review, we report 23 plants that have been used for cancer management in Africa, where the anticancer extracts are usually prepared from barks, fruits, leaves, roots, and stems of these plants. Extensive information is reported about the bioactive compounds present in these plants as well as their potential activities against various forms of cancer. However, information on the anticancer properties of other African medicinal plants is insufficient. Therefore, there is a need to isolate and evaluate the anticancer potential of bioactive compounds from other African medicinal plants. Further studies on these plants will allow the elucidation of their anticancer mechanisms of action and allow the identification of phytochemicals that are responsible for their anticancer properties. Overall, this review provides consolidated and extensive information not only on diverse medicinal plants of Africa but on the different types of cancer that these plants are used to manage and the diverse mechanisms and pathways that are involved during cancer alleviation.
Collapse
Affiliation(s)
- Goabaone Gaobotse
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana,*Correspondence: Goabaone Gaobotse, ; Kabo Masisi, ; Abdullah Makhzoum,
| | - Srividhya Venkataraman
- Virology Laboratory, Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Phenyo D. Brown
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana
| | - Kabo Masisi
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana,*Correspondence: Goabaone Gaobotse, ; Kabo Masisi, ; Abdullah Makhzoum,
| | - Tebogo E. Kwape
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana
| | - David O. Nkwe
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana
| | - Gaolathe Rantong
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana
| | - Abdullah Makhzoum
- Department of Biological Sciences and Biotechnology, Faculty of Sciences, Botswana International University of Science and Technology, Palapye, Botswana,*Correspondence: Goabaone Gaobotse, ; Kabo Masisi, ; Abdullah Makhzoum,
| |
Collapse
|
11
|
Zhang Z, Huang Y, Li J, Su F, Kuo JC, Hu Y, Zhao X, Lee RJ. Antitumor Activity of Anti-miR-21 Delivered through Lipid Nanoparticles. Adv Healthc Mater 2023; 12:e2202412. [PMID: 36412002 PMCID: PMC11468686 DOI: 10.1002/adhm.202202412] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/17/2022] [Indexed: 11/23/2022]
Abstract
The ability of lipid nanoparticles (LNPs) to deliver nucleic acids have shown a great therapeutic potential to treat a variety of diseases. Here, an optimized formulation of QTsome lipid nanoparticles (QTPlus) is utilized to deliver an anti-miR-21 (AM21) against cancer. The miR-21 downstream gene regulation and antitumor activity is evaluated using mouse and human cancer cells and macrophages. The antitumor activity of QTPlus encapsulating AM21 (QTPlus-AM21) is further evaluated in combination with erlotinib and atezolizumab (ATZ). QTPlus-AM21 demonstrates a superior miR-21-dependent gene regulation and eventually inhibits A549 non-small cell lung cancer growth in vitro. QTPlus-AM21 further induces chemo-sensitization of A549 cells to erlotinib with a combination index of 0.6 in inhibiting A549 cell growth. When systemically administers to MC38 tumor-bearing mouse model, QTPlus-AM21 exhibits an antitumor immune response with over 80% tumor growth inhibition (TGI%) and over twofold and fourfold PD-1 and PD-L1 upregulation in tumors and spleens. The combination therapy of QTPlus-AM21 and ATZ further shows a higher antitumor response (TGI% over 90%) and successfully increases M1 macrophages and CD8 T cells into TME. This study provides new insights into the antitumor mechanism of AM21 and shows great promise of QTPlus-AM21 in combination with chemotherapies and immunotherapies.
Collapse
Affiliation(s)
- Zhongkun Zhang
- Division of Pharmaceutics and PharmacologyCollege of PharmacyThe Ohio State University500 W 12th AvenueColumbusOH43210USA
| | - Yirui Huang
- Division of Pharmaceutics and PharmacologyCollege of PharmacyThe Ohio State University500 W 12th AvenueColumbusOH43210USA
| | - Jing Li
- Zhejiang Haichang Biotechnology Co., Ltd.HangzhouZhejiang310000P. R. China
| | - Fei Su
- Zhejiang Haichang Biotechnology Co., Ltd.HangzhouZhejiang310000P. R. China
| | - Jimmy Chun‐Tien Kuo
- Division of Pharmaceutics and PharmacologyCollege of PharmacyThe Ohio State University500 W 12th AvenueColumbusOH43210USA
| | - Yingwen Hu
- The Whiteoak Group, Inc.RockvilleMD20855USA
| | | | - Robert J. Lee
- Division of Pharmaceutics and PharmacologyCollege of PharmacyThe Ohio State University500 W 12th AvenueColumbusOH43210USA
| |
Collapse
|
12
|
Rahimi H, Abdollahzade A, Ramezani M, Alibolandi M, Abnous K, Taghdisi SM. Targeted delivery of doxorubicin to tumor cells using engineered circular bivalent aptamer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
13
|
Kara G, Arun B, Calin GA, Ozpolat B. miRacle of microRNA-Driven Cancer Nanotherapeutics. Cancers (Basel) 2022; 14:3818. [PMID: 35954481 PMCID: PMC9367393 DOI: 10.3390/cancers14153818] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/28/2022] [Accepted: 08/04/2022] [Indexed: 12/23/2022] Open
Abstract
MicroRNAs (miRNAs) are non-protein-coding RNA molecules 20-25 nucleotides in length that can suppress the expression of genes involved in numerous physiological processes in cells. Accumulating evidence has shown that dysregulation of miRNA expression is related to the pathogenesis of various human diseases and cancers. Thus, stragegies involving either restoring the expression of tumor suppressor miRNAs or inhibiting overexpressed oncogenic miRNAs hold potential for targeted cancer therapies. However, delivery of miRNAs to tumor tissues is a challenging task. Recent advances in nanotechnology have enabled successful tumor-targeted delivery of miRNA therapeutics through newly designed nanoparticle-based carrier systems. As a result, miRNA therapeutics have entered human clinical trials with promising results, and they are expected to accelerate the transition of miRNAs from the bench to the bedside in the next decade. Here, we present recent perspectives and the newest developments, describing several engineered natural and synthetic novel miRNA nanocarrier formulations and their key in vivo applications and clinical trials.
Collapse
Affiliation(s)
- Goknur Kara
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Chemistry, Biochemistry Division, Ordu University, Ordu 52200, Turkey
| | - Banu Arun
- Department of Breast Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
| | - George A. Calin
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
| | - Bulent Ozpolat
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
14
|
Zhang X, Rotllan N, Canfrán-Duque A, Sun J, Toczek J, Moshnikova A, Malik S, Price NL, Araldi E, Zhong W, Sadeghi MM, Andreev OA, Bahal R, Reshetnyak YK, Suárez Y, Fernández-Hernando C. Targeted Suppression of miRNA-33 Using pHLIP Improves Atherosclerosis Regression. Circ Res 2022; 131:77-90. [PMID: 35534923 PMCID: PMC9640270 DOI: 10.1161/circresaha.121.320296] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 05/03/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND miRNA therapeutics have gained attention during the past decade. These oligonucleotide treatments can modulate the expression of miRNAs in vivo and could be used to correct the imbalance of gene expression found in human diseases such as obesity, metabolic syndrome, and atherosclerosis. The in vivo efficacy of current anti-miRNA technologies hindered by physiological and cellular barriers to delivery into targeted cells and the nature of miRNAs that allows one to target an entire pathway that may lead to deleterious off-target effects. For these reasons, novel targeted delivery systems to inhibit miRNAs in specific tissues will be important for developing effective therapeutic strategies for numerous diseases including atherosclerosis. METHODS We used pH low-insertion peptide (pHLIP) constructs as vehicles to deliver microRNA-33-5p (miR-33) antisense oligonucleotides to atherosclerotic plaques. Immunohistochemistry and histology analysis was performed to assess the efficacy of miR-33 silencing in atherosclerotic lesions. We also assessed how miR-33 inhibition affects gene expression in monocytes/macrophages by single-cell RNA transcriptomics. RESULTS The anti-miR-33 conjugated pHLIP constructs are preferentially delivered to atherosclerotic plaque macrophages. The inhibition of miR-33 using pHLIP-directed macrophage targeting improves atherosclerosis regression by increasing collagen content and decreased lipid accumulation within vascular lesions. Single-cell RNA sequencing analysis revealed higher expression of fibrotic genes (Col2a1, Col3a1, Col1a2, Fn1, etc) and tissue inhibitor of metalloproteinase 3 (Timp3) and downregulation of Mmp12 in macrophages from atherosclerotic lesions targeted by pHLIP-anti-miR-33. CONCLUSIONS This study provides proof of principle for the application of pHLIP for treating advanced atherosclerosis via pharmacological inhibition of miR-33 in macrophages that avoid the deleterious effects in other metabolic tissues. This may open new therapeutic opportunities for atherosclerosis-associated cardiovascular diseases via selective delivery of other protective miRNAs.
Collapse
Affiliation(s)
- Xinbo Zhang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Alberto Canfrán-Duque
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jonathan Sun
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jakub Toczek
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Section of Cardiology, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Anna Moshnikova
- Department Physics, University of Rhode Island, Kingston, Rhode Island, USA
| | - Shipra Malik
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Nathan L. Price
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Elisa Araldi
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Wen Zhong
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mehran M. Sadeghi
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Cardiovascular Molecular Imaging Laboratory, Section of Cardiovascular Medicine and Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Section of Cardiology, Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Oleg A. Andreev
- Department Physics, University of Rhode Island, Kingston, Rhode Island, USA
| | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Yana K. Reshetnyak
- Department Physics, University of Rhode Island, Kingston, Rhode Island, USA
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Integrative Cell Signaling and Neurobiology of Metabolism Program, Department of Comparative Medicine and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
15
|
Zhang C, Ma Y, Zhang J, Kuo JCT, Zhang Z, Xie H, Zhu J, Liu T. Modification of Lipid-Based Nanoparticles: An Efficient Delivery System for Nucleic Acid-Based Immunotherapy. Molecules 2022; 27:molecules27061943. [PMID: 35335310 PMCID: PMC8949521 DOI: 10.3390/molecules27061943] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Lipid-based nanoparticles (LBNPs) are biocompatible and biodegradable vesicles that are considered to be one of the most efficient drug delivery platforms. Due to the prominent advantages, such as long circulation time, slow drug release, reduced toxicity, high transfection efficiency, and endosomal escape capacity, such synthetic nanoparticles have been widely used for carrying genetic therapeutics, particularly nucleic acids that can be applied in the treatment for various diseases, including congenital diseases, cancers, virus infections, and chronic inflammations. Despite great merits and multiple successful applications, many extracellular and intracellular barriers remain and greatly impair delivery efficacy and therapeutic outcomes. As such, the current state of knowledge and pitfalls regarding the gene delivery and construction of LBNPs will be initially summarized. In order to develop a new generation of LBNPs for improved delivery profiles and therapeutic effects, the modification strategies of LBNPs will be reviewed. On the basis of these developed modifications, the performance of LBNPs as therapeutic nanoplatforms have been greatly improved and extensively applied in immunotherapies, including infectious diseases and cancers. However, the therapeutic applications of LBNPs systems are still limited due to the undesirable endosomal escape, potential aggregation, and the inefficient encapsulation of therapeutics. Herein, we will review and discuss recent advances and remaining challenges in the development of LBNPs for nucleic acid-based immunotherapy.
Collapse
Affiliation(s)
- Chi Zhang
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Z.Z.)
| | - Yifan Ma
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; (Y.M.); (J.Z.)
| | - Jingjing Zhang
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA; (Y.M.); (J.Z.)
| | - Jimmy Chun-Tien Kuo
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Z.Z.)
| | - Zhongkun Zhang
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (C.Z.); (J.C.-T.K.); (Z.Z.)
| | - Haotian Xie
- Department of Statistics, The Ohio State University, Columbus, OH 43210, USA;
| | - Jing Zhu
- College of Nursing and Health Innovation, The University of Texas Arlington, Arlington, TX 76010, USA
- Correspondence: (J.Z.); (T.L.); Tel.: +1-614-570-1164 (J.Z.); +86-186-6501-3854 (T.L.)
| | - Tongzheng Liu
- College of Pharmacy, Jinan University, Guangzhou 511443, China
- Correspondence: (J.Z.); (T.L.); Tel.: +1-614-570-1164 (J.Z.); +86-186-6501-3854 (T.L.)
| |
Collapse
|
16
|
Wu L, Zhou W, Lin L, Chen A, Feng J, Qu X, Zhang H, Yue J. Delivery of therapeutic oligonucleotides in nanoscale. Bioact Mater 2022; 7:292-323. [PMID: 34466734 PMCID: PMC8379367 DOI: 10.1016/j.bioactmat.2021.05.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/28/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
Therapeutic oligonucleotides (TOs) represent one of the most promising drug candidates in the targeted cancer treatment due to their high specificity and capability of modulating cellular pathways that are not readily druggable. However, efficiently delivering of TOs to cancer cellular targets is still the biggest challenge in promoting their clinical translations. Emerging as a significant drug delivery vector, nanoparticles (NPs) can not only protect TOs from nuclease degradation and enhance their tumor accumulation, but also can improve the cell uptake efficiency of TOs as well as the following endosomal escape to increase the therapeutic index. Furthermore, targeted and on-demand drug release of TOs can also be approached to minimize the risk of toxicity towards normal tissues using stimuli-responsive NPs. In the past decades, remarkable progresses have been made on the TOs delivery based on various NPs with specific purposes. In this review, we will first give a brief introduction on the basis of TOs as well as the action mechanisms of several typical TOs, and then describe the obstacles that prevent the clinical translation of TOs, followed by a comprehensive overview of the recent progresses on TOs delivery based on several various types of nanocarriers containing lipid-based nanoparticles, polymeric nanoparticles, gold nanoparticles, porous nanoparticles, DNA/RNA nanoassembly, extracellular vesicles, and imaging-guided drug delivery nanoparticles.
Collapse
Affiliation(s)
- Lei Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Wenhui Zhou
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Lihua Lin
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Anhong Chen
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Jing Feng
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Xiangmeng Qu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory and Turku Bioscience Centre, Åbo Akademi University, Turku, 20520, Finland
| | - Jun Yue
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| |
Collapse
|
17
|
Ma X, Wang X, Liu C, Ge B, He H, Dai Q, Zhang Z, Yu J, Nau WM, Huang F. Self-assembled theranostic microcarrier targeting tumor cells with high metastatic potential. MATERIALS & DESIGN 2021; 212:110196. [DOI: 10.1016/j.matdes.2021.110196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
miRNA Delivery by Nanosystems: State of the Art and Perspectives. Pharmaceutics 2021; 13:pharmaceutics13111901. [PMID: 34834316 PMCID: PMC8619868 DOI: 10.3390/pharmaceutics13111901] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/25/2021] [Accepted: 11/04/2021] [Indexed: 01/09/2023] Open
Abstract
MicroRNAs (miRNAs) are short (~21-23 nucleotides), non-coding endogenous RNA molecules that modulate gene expression at the post-transcriptional level via the endogenous RNA interference machinery of the cell. They have emerged as potential biopharmaceuticals candidates for the treatment of various diseases, including cancer, cardiovascular and metabolic diseases. However, in order to advance miRNAs therapeutics into clinical settings, their delivery remains a major challenge. Different types of vectors have been investigated to allow the delivery of miRNA in the diseased tissue. In particular, non-viral delivery systems have shown important advantages such as versatility, low cost, easy fabrication and low immunogenicity. Here, we present a general overview of the main types of non-viral vectors developed for miRNA delivery, with their advantages, limitations and future perspectives.
Collapse
|
19
|
Ward DM, Shodeinde AB, Peppas NA. Innovations in Biomaterial Design toward Successful RNA Interference Therapy for Cancer Treatment. Adv Healthc Mater 2021; 10:e2100350. [PMID: 33973393 PMCID: PMC8273125 DOI: 10.1002/adhm.202100350] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/27/2021] [Indexed: 12/11/2022]
Abstract
Gene regulation using RNA interference (RNAi) therapy has been developed as one of the frontiers in cancer treatment. The ability to tailor the expression of genes by delivering synthetic oligonucleotides to tumor cells has transformed the way scientists think about treating cancer. However, its clinical application has been limited due to the need to deliver synthetic RNAi oligonucleotides efficiently and effectively to target cells. Advances in nanotechnology and biomaterials have begun to address the limitations to RNAi therapeutic delivery, increasing the likelihood of RNAi therapeutics for cancer treatment in clinical settings. Herein, innovations in the design of nanocarriers for the delivery of oligonucleotides for successful RNAi therapy are discussed.
Collapse
Affiliation(s)
- Deidra M Ward
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Aaliyah B Shodeinde
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, 78712, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, 78712, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave. Stop A1900, Austin, TX, 78712, USA
- Department of Pediatrics and Department of Surgery and Perioperative Care, Dell Medical School, 1601 Trinity St., Bldg. B, Stop Z0800, Austin, TX, 78712, USA
| |
Collapse
|
20
|
Li D, Gao C, Kuang M, Xu M, Wang B, Luo Y, Teng L, Xie J. Nanoparticles as Drug Delivery Systems of RNAi in Cancer Therapy. Molecules 2021; 26:2380. [PMID: 33921892 PMCID: PMC8073355 DOI: 10.3390/molecules26082380] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/26/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
RNA interference (RNAi) can mediate gene-silencing by knocking down the expression of a target gene via cellular machinery with much higher efficiency in contrast to other antisense-based approaches which represents an emerging therapeutic strategy for combating cancer. Distinct characters of nanoparticles, such as distinctive size, are fundamental for the efficient delivery of RNAi therapeutics, allowing for higher targeting and safety. In this review, we present the mechanism of RNAi and briefly describe the hurdles and concerns of RNAi as a cancer treatment approach in systemic delivery. Furthermore, the current nanovectors for effective tumor delivery of RNAi therapeutics are classified, and the characteristics of different nanocarriers are summarized.
Collapse
Affiliation(s)
- Diedie Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Chengzhi Gao
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Meiyan Kuang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Minhao Xu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Ben Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Yi Luo
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun 130012, China;
| | - Jing Xie
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; (D.L.); (C.G.); (M.K.); (M.X.); (B.W.); (Y.L.)
| |
Collapse
|
21
|
Wang D, Wang X, Wang L, Zhang J, Ma J, Xia G, Hong B. Antisense microRNA185 loaded liposome for efficient inhibition of the hepatic endogenous microRNA185 level. Eur J Pharm Sci 2021; 161:105803. [PMID: 33722732 DOI: 10.1016/j.ejps.2021.105803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/08/2021] [Accepted: 03/07/2021] [Indexed: 02/06/2023]
Abstract
MicroRNA185 (miR185), an endogenous noncoding RNA with 23 nucleotides, is one of key posttranscriptional modulators of cholesterol metabolism in hepatic cells. The antisense inhibitor of miR185 (miR185i) could decrease cholesterol level in vivo, providing a promising agent for anti-atherosclerosis strategy. In this work, a novel LipomiR185i was constructed by thin film hydration method and post-PEGylation as DOPE: DOTAP: Chol: DSPE-PEG2000 at the molar ratio of 1:1:1:0.1 with a nitrogen-to-phosphate ratio of 3, through the optimization of three cationic lipids (DOTAP, DODMA and DLin-MC3-DMA), six helper lipids (PC-98T, HSPC, DOPE, DMPC, DPPC and DSPC), different amounts and incorporation approaches of DSPE-PEG2000 and nitrogen-to-phosphate ratio. LipomiR185i was characterized with a particle size of 174 ± 11 nm, a zeta potential of 7.0 ± 3.3 mV, high encapsulation efficiency and transfection activity. It could protect miR185i from the rapid degradation by nucleases in serum, enhance cellular uptake and promote lysosomal escape in HepG2 cells. LipomiR185i could accumulate in the liver and remain for at least two weeks. More importantly, LipomiR185i significantly down-regulated the hepatic endogenous miR185 level in vitro and in vivo without significant tissue damage at 14 mg⋅kg-1. The construction of LipomiR185i provides a potential anti-atherosclerotic nanodrug as well as a platform for delivering small RNAs to the liver efficiently and safely.
Collapse
Affiliation(s)
- Dan Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xuelei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Li Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Jin Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jie Ma
- Department of Biotherapy, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Bin Hong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing 100050, China.
| |
Collapse
|
22
|
Ding X, Zheng J, Cao M. Circ_0004771 Accelerates Cell Carcinogenic Phenotypes via Suppressing miR-1253-Mediated DDAH1 Inhibition in Breast Cancer. Cancer Manag Res 2021; 13:1-11. [PMID: 33442289 PMCID: PMC7797298 DOI: 10.2147/cmar.s273783] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/04/2020] [Indexed: 12/20/2022] Open
Abstract
Background Circ_0004771 was demonstrated to mediate cell growth promotion and apoptosis suppression in breast cancer (BC). Herein, the precise functions and mechanism of circ_0004771 in the biological property of BC cells were investigated. Methods The expression of circ_0004771, microRNA (miR)-1253 and dimethylarginine dimethylaminohydrolase 1 (DDAH1) mRNA was analyzed using quantitative real-time polymerase chain reaction. The proliferation, apoptosis, migration, invasion, adhesion, Western blot and in vivo tumorigenesis assays were employed to evaluate the roles of circ_0004771 and DDAH1 in BC tumorigenesis. The interaction between miR-1253 and circ_0004771 or DDAH1 was validated by dual-luciferase reporter, pull-down and RNA immunoprecipitation (RIP) assay. Exosomes were isolated by Exoquick-TC® methods, and qualified using Nanosight™ technology and Western blot. Results Circ_0004771 or DDAH1 expression was elevated in BC, and silencing either of them suppressed cell malignant phenotypes, thus impeding BC progression. Importantly, circ_0004771 up-regulation attenuated the anticancer action of DDAH1-knockdown in BC. Additionally, we confirmed that circ_0004771 functioned as a sponge of miR-1253 to up-regulate DDAH1 expression. Moreover, xenograft assay exhibited that circ_0004771 knockdown also hindered tumor growth in vivo via regulating DDAH1 and miR-1253. Besides that, it was found that circ_0004771 was packaged into exosomes isolated from the serum of BC. Conclusion Circ_0004771 accelerated cell carcinogenic phenotypes via up-regulating DDAH1 expression through absorbing miR-1253 in BC. Besides, circ_0004771 was packaged into exosomes isolated from the serum of BC. All these findings suggested a promising molecular target for BC treatment.
Collapse
Affiliation(s)
- Xubei Ding
- Thyroid and Breast Surgery, Jingmen No.1 People's Hospital, Jingmen, Hubei, People's Republic of China
| | - Junjun Zheng
- Pharmacy Intravenous Admixture Services, Jingmen No.2 People's Hospital, Jingmen, Hubei, People's Republic of China
| | - Mingxiang Cao
- Department of Anesthesiology, Jingmen No.1 People's Hospital, Jingmen, Hubei, People's Republic of China
| |
Collapse
|
23
|
Trittmann JK, Almazroue H, Jin Y, Nelin LD. DDAH1 regulates apoptosis and angiogenesis in human fetal pulmonary microvascular endothelial cells. Physiol Rep 2020; 7:e14150. [PMID: 31209995 PMCID: PMC6579941 DOI: 10.14814/phy2.14150] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 01/17/2023] Open
Abstract
Nitric Oxide (NO) is an endogenous pulmonary vasodilator produced by endothelial NO synthase (eNOS). Asymmetric dimethyl L‐arginine (ADMA) is an endogenous inhibitor of eNOS activity. In endothelial cells, ADMA is hydrolyzed to L‐citrulline primarily by dimethylarginine dimethyl‐aminohydrolase‐1 (DDAH1). We tested the hypothesis that DDAH1 expression is essential for maintaining NO production in human fetal pulmonary microvascular endothelial cells (hfPMVEC), such that knockdown of DDAH1 expression will lead to decreased NO production resulting in less caspase‐3 activation and less tube formation. We found that hfPMVEC transfected with DDAH1 siRNA had lower NO production than control, with no difference in eNOS protein levels between groups. hfPMVEC transfected with DDAH1 siRNA had lower protein levels of cleaved caspase‐3 and ‐8 than control. Both DDAH1 siRNA‐ and ADMA‐treated hfPMVEC had greater numbers of viable cells than controls. Angiogenesis was assessed using tube formation assays in matrigel, and tube formation was lower after either DDAH1 siRNA transfection or ADMA treatment than controls. Addition of an NO donor restored cleaved caspase‐3 and ‐8 protein levels after DDAH1 siRNA transfection in hfPMVEC to essentially the levels seen in scramble control. Addition of a putative caspase‐3 inhibitor to DDAH1 siRNA transfected and NO‐donor treated cells led to greater numbers of viable cells and far less angiogenesis than in any other group studied. We conclude that in hfPMVEC, DDAH1 is central to the regulation of NO‐mediated caspase‐3 activation and the resultant apoptosis and angiogenesis. Our findings suggest that DDAH1 may be a potential therapeutic target in pulmonary hypertensive disorders.
Collapse
Affiliation(s)
- Jennifer K Trittmann
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Hanadi Almazroue
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
24
|
Boca S, Gulei D, Zimta AA, Onaciu A, Magdo L, Tigu AB, Ionescu C, Irimie A, Buiga R, Berindan-Neagoe I. Nanoscale delivery systems for microRNAs in cancer therapy. Cell Mol Life Sci 2020; 77:1059-1086. [PMID: 31637450 PMCID: PMC11105078 DOI: 10.1007/s00018-019-03317-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/26/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022]
Abstract
Concomitant with advances in research regarding the role of miRNAs in sustaining carcinogenesis, major concerns about their delivery options for anticancer therapies have been raised. The answer to this problem may come from the world of nanoparticles such as liposomes, exosomes, polymers, dendrimers, mesoporous silica nanoparticles, quantum dots and metal-based nanoparticles which have been proved as versatile and valuable vehicles for many biomolecules including miRNAs. In another train of thoughts, the general scheme of miRNA modulation consists in inhibition of oncomiRNA expression and restoration of tumor suppressor ones. The codelivery of two miRNAs or miRNAs in combination with chemotherapeutics or small molecules was also proposed. The present review presents the latest advancements in miRNA delivery based on nanoparticle-related strategies.
Collapse
Affiliation(s)
- Sanda Boca
- Nanobiophotonics and Laser Microspectroscopy Center, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, 42 T. Laurian, 400271, Cluj-Napoca, Romania
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Diana Gulei
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Anca Onaciu
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Lorand Magdo
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania
| | - Adrian Bogdan Tigu
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Calin Ionescu
- 5th Surgical Department, Municipal Hospital, Cluj-Napoca, Romania
- "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandru Irimie
- Department of Oncological Surgery and Gynecological Oncology, 400015, Cluj-Napoca, Romania
- Department of Surgery, The Oncology Institute "Prof. Dr. Ion Chiricuta", 400015, Cluj-Napoca, Romania
| | - Rares Buiga
- Department of Pathology, "Prof Dr. Ion Chiricuta" Oncology Institute, Cluj-Napoca, Romania.
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu", University of Medicine and Pharmacy, 23 Marinescu Street, 400337, Cluj-Napoca, Romania.
- MEDFUTURE-Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy, 23 Marinescu Street, Cluj-Napoca, Romania.
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", 34-36 Republicii Street, Cluj-Napoca, Romania.
| |
Collapse
|
25
|
Abnous K, Danesh NM, Ramezani M, Alibolandi M, Bahreyni A, Lavaee P, Moosavian SA, Taghdisi SM. A smart ATP-responsive chemotherapy drug-free delivery system using a DNA nanostructure for synergistic treatment of breast cancer in vitro and in vivo. J Drug Target 2020; 28:852-859. [PMID: 31916879 DOI: 10.1080/1061186x.2020.1712407] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This study demonstrated a chemotherapy drug-free delivery system for breast cancer treatment based on a simple DNA nanostructure composed of sequence 1 containing ATP and AS1411 aptamers and sequence 2 containing antimiR-21. The DNA nanostructure was used for co-delivery of KLA peptide and antimiR-21 as antiapoptotic agents. These therapeutic agents could not be internalised into eukaryotic cells freely which is one of the great features of this targeting platform. The presented delivery system was ATP-responsive, leading to disassembly of the DNA nanostructure in high ATP concentration of cancer cells and restoration of the function of antimiR-21 in these cells. The DNA nanostructure was associated with high cellular uptake by MCF-7 and 4T1 cells due to expression of nucleolin as target of AS1411 on their plasma membranes, while the developed targeting platform could not be internalised into CHO cells because of lack of the active targeting moiety on their surfaces. Furthermore, the results showed that co-delivery of antimiR-21 and KLA peptide using the DNA nanostructure could efficiently prohibit tumour growth in vitro and in vivo and induce a synergistic anticancer activity. Thus, this work provides a new ATP-responsive nanotargeting delivery system and synergistic chemotherapy drug-free regimen for cancer treatment.
Collapse
Affiliation(s)
- Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Bahreyni
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Parirokh Lavaee
- Academic Center for Education, Culture and Research, Research Institute for Industrial Biotechnology, Industrial Biotechnology on Microorganisms, Mashhad, Iran
| | - Seyedeh Alia Moosavian
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Peng Y, Bariwal J, Kumar V, Tan C, Mahato RI. Organic Nanocarriers for Delivery and Targeting of Therapeutic Agents for Cancer Treatment. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900136] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yang Peng
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Jitender Bariwal
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Virender Kumar
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug DeliveryUniversity of Mississippi University MS 38677 USA
| | - Ram I. Mahato
- Department of Pharmaceutical SciencesUniversity of Nebraska Medical Center Omaha NE 68198 USA
| |
Collapse
|
27
|
Petrek H, Yu A. MicroRNAs in non-small cell lung cancer: Gene regulation, impact on cancer cellular processes, and therapeutic potential. Pharmacol Res Perspect 2019; 7:e00528. [PMID: 31859460 PMCID: PMC6923806 DOI: 10.1002/prp2.528] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/29/2022] Open
Abstract
Lung cancer remains the most lethal cancer among men and women in the United States and worldwide. The majority of lung cancer cases are classified as non-small cell lung cancer (NSCLC). Developing new therapeutics on the basis of better understanding of NSCLC biology is critical to improve the treatment of NSCLC. MicroRNAs (miRNAs or miRs) are a superfamily of genome-derived, small noncoding RNAs that govern posttranscriptional gene expression in cells. Functional miRNAs are commonly dysregulated in NSCLC, caused by genomic deletion, methylation, or altered processing, which may lead to the changes of many cancer-related pathways and processes, such as growth and death signaling, metabolism, angiogenesis, cell cycle, and epithelial to mesenchymal transition, as well as sensitivity to current therapies. With the understanding of miRNA biology in NSCLC, there are growing interests in developing new therapeutic strategies, namely restoration of tumor suppressive miRNAs and inhibition of tumor promotive miRNAs, to combat against NSCLC. In this article, we provide an overview on the molecular features of NSCLC and current treatment options with a focus on pharmacotherapy and personalized medicine. By illustrating the roles of miRNAs in the control of NSCLC tumorigenesis and progression, we highlight the latest efforts in assessing miRNA-based therapies in animal models and discuss some critical challenges in developing RNA therapeutics.
Collapse
Affiliation(s)
- Hannah Petrek
- Department of Biochemistry & Molecular MedicineUC Davis School of MedicineSacramentoCAUSA
| | - Ai‐Ming Yu
- Department of Biochemistry & Molecular MedicineUC Davis School of MedicineSacramentoCAUSA
| |
Collapse
|
28
|
Scheideler M, Vidakovic I, Prassl R. Lipid nanocarriers for microRNA delivery. Chem Phys Lipids 2019; 226:104837. [PMID: 31689410 DOI: 10.1016/j.chemphyslip.2019.104837] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023]
Abstract
Non-coding RNAs (ncRNAs) like microRNAs (miRNAs) or small interference RNAs (siRNAs) with their power to selectively silence any gene of interest enable the targeting of so far 'undruggable' proteins and diseases. Such RNA molecules have gained much attention from biotech and pharmaceutical companies, which led to the first Food and Drug Administration (FDA) approved ncRNA therapeutic in 2018. However, the main barrier in clinical practice of ncRNAs is the lack of an effective delivery system that can protect the RNA molecules from nuclease degradation, deliver them to specific tissues and cell types, and release them into the cytoplasm of the targeted cells, all without inducing adverse effects. For that reason, drug delivery approaches, formulations, technologies and systems for transporting pharmacological ncRNA compounds to achieve a diagnostic or therapeutic effect in the human body are in demand. Here, we review the development of therapeutic lipid-based nanoparticles for delivery of miRNAs, one class of endogenous ncRNAs with specific regulatory functions. We outline challenges and opportunities for advanced miRNA-based therapies, and discuss the complexity associated with the delivery of functional miRNAs. Novel strategies are addressed how to deal with the most critical points in miRNA delivery, such as toxicity, specific targeting of disease sites, proper cellular uptake and endosomal escape of miRNAs. Current fields of application and various preclinical settings involving miRNA therapeutics are discussed, providing an outlook to future clinical approaches. Following the current trends and technological developments in nanomedicine exciting new delivery systems for ncRNA-based therapeutics can be expected in upcoming years.
Collapse
Affiliation(s)
- Marcel Scheideler
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Ivan Vidakovic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria.
| | - Ruth Prassl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria.
| |
Collapse
|
29
|
Bertucci A, Kim KH, Kang J, Zuidema JM, Lee SH, Kwon EJ, Kim D, Howell SB, Ricci F, Ruoslahti E, Jang HJ, Sailor MJ. Tumor-Targeting, MicroRNA-Silencing Porous Silicon Nanoparticles for Ovarian Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:23926-23937. [PMID: 31251556 DOI: 10.1021/acsami.9b07980] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Silencing of aberrantly expressed microRNAs (miRNAs or miRs) has emerged as one of the strategies for molecular targeted cancer therapeutics. In particular, miR-21 is an oncogenic miRNA overexpressed in many tumors, including ovarian cancer. To achieve efficient administration of anti-miR therapeutics, delivery systems are needed that can ensure local accumulation in the tumor environment, low systemic toxicity, and reduced adverse side effects. In order to develop an improved anti-miR therapeutic agent for the treatment of ovarian cancer, a nanoformulation is engineered that leverages biodegradable porous silicon nanoparticles (pSiNPs) encapsulating an anti-miR-21 locked nucleic acid payload and displaying a tumor-homing peptide for targeted distribution. Targeting efficacy, miR-21 silencing, and anticancer activity are optimized in vitro on a panel of ovarian cancer cell lines, and a formulation of anti-miR-21 in a pSiNP displaying the targeting peptide CGKRK is identified for in vivo evaluation. When this nanoparticulate agent is delivered to mice bearing tumor xenografts, a substantial inhibition of tumor growth is achieved through silencing of miR-21. This study presents the first successful application of tumor-targeted anti-miR porous silicon nanoparticles for the treatment of ovarian cancer in a mouse xenograft model.
Collapse
Affiliation(s)
- Alessandro Bertucci
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Rome , 00133 , Italy
| | | | | | | | | | | | | | | | - Francesco Ricci
- Department of Chemical Sciences and Technologies , University of Rome Tor Vergata , Rome , 00133 , Italy
| | - Erkki Ruoslahti
- Cancer Center , Sanford Burnham Prebys Medical Discovery Institute , La Jolla , California 92037 , United States
| | | | | |
Collapse
|
30
|
Dua K, Chellappan DK, Singhvi G, de Jesus Andreoli Pinto T, Gupta G, Hansbro PM. Targeting microRNAs using nanotechnology in pulmonary diseases. Panminerva Med 2019; 60:230-231. [PMID: 30563304 DOI: 10.23736/s0031-0808.18.03459-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia - .,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, Australia - .,The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, Australia -
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Malaysia
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani, India
| | | | - Gaurav Gupta
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, India
| | - Philip M Hansbro
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, Australia.,The Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, Australia
| |
Collapse
|
31
|
Yu J, Peng J, Luan Z, Zheng F, Su W. MicroRNAs as a Novel Tool in the Diagnosis of Liver Lipid Dysregulation and Fatty Liver Disease. Molecules 2019; 24:molecules24020230. [PMID: 30634538 PMCID: PMC6358728 DOI: 10.3390/molecules24020230] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/23/2018] [Accepted: 12/24/2018] [Indexed: 02/07/2023] Open
Abstract
In recent years, metabolic disorder, especially fatty liver disease, has been considered a major challenge to global health. The attention of researchers focused on expanding knowledge of the regulation mechanism behind these diseases and towards the new diagnostics tools and treatments. The pathophysiology of the fatty liver disease is undoubtedly complex. Abnormal hepatic lipid accumulation is a major symptom of most metabolic diseases. Therefore, the identification of novel regulation factors of lipid metabolism is important and meaningful. As a new diagnostic tool, the function of microRNAs during fatty liver disease has recently come into notice in biological research. Accumulating evidence supports the influence of miRNAs in lipid metabolism. In this review, we discuss the potential role of miRNAs in liver lipid metabolism and the pathogenesis of fatty liver disease.
Collapse
Affiliation(s)
- Jingwei Yu
- Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen 518060, China.
- Department of Biology, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Jun Peng
- Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen 518060, China.
| | - Zhilin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Feng Zheng
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Wen Su
- Shenzhen University Medical Center, Shenzhen University Health Science Center, Shenzhen 518060, China.
| |
Collapse
|
32
|
Chen Y, Sun J, Huang Y, Liu Y, Liang L, Yang D, Lu B, Li S. Targeted codelivery of doxorubicin and IL-36γ expression plasmid for an optimal chemo-gene combination therapy against cancer lung metastasis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 15:129-141. [PMID: 30308300 DOI: 10.1016/j.nano.2018.09.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/07/2018] [Accepted: 09/19/2018] [Indexed: 10/28/2022]
Abstract
Cancer metastasis is the main cause for the high mortality in breast cancer patients. In this work we developed a polymer POEG-st-Pmor for targeted co-delivery of IL-36γ expression plasmid and doxorubicin (Dox) to lung metastasis of breast cancer. The polymer readily formed micelles that were effective in loading Dox and simultaneously forming complexes with IL-36γ plasmid. Interestingly, particles co-loaded with Dox and plasmid was significantly smaller and more stable than the particles loaded with Dox only. Gene transfection in both lungs and s.c. tumors was significantly higher with our polymer compared to PEI. In addition, the Dox + IL-36γ/POEG-st-Pmor not only could bring improved anti-metastatic effect but synergistically enhance the type I immune response by increasing the IFN-γ positive CD4+ and CD8+ T cells and simultaneously decreasing the immunosuppressive myeloid-derived suppressor cells in the lung. POEG-st-Pmor may represent a simple and effective delivery system for an optimal chemo-gene combination therapy.
Collapse
Affiliation(s)
- Yichao Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jingjing Sun
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Lei Liang
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Da Yang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
33
|
Yang X, Zhang L, Song X, He W, Zhang D, Lu Q, Wu J, Wu C, Jiang J. MicroRNA-613 promotes colon cancer cell proliferation, invasion and migration by targeting ATOH1. Biochem Biophys Res Commun 2018; 504:827-833. [PMID: 30219232 DOI: 10.1016/j.bbrc.2018.09.054] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 09/09/2018] [Indexed: 02/01/2023]
Abstract
The aim of the present study is to investigate the expression and function of miR-613 in colon cancer (CC) and illuminate the molecular mechanisms underlying miR-613-regulated CC progression. Our data demonstrated that miR-613 was upregulated in CC tissue samples (P = 0.009) and human CC cell lines (HCT-116 and Lovo; P = 0.001 and P = 0.003, respectively), which also promoted the proliferation, invasion and migration of CC cells (P < 0.05). The dual-luciferase reporter assay confirmed that Atonal homolog1 (ATOH1) was the target mRNA of miR-613. Rescue experiments showed that ATOH1 overexpression vector significantly reversed the stimulative effects of miR-613 mimic on the progression of HCT-116 and Lovo cells (P < 0.001). Positive ATOH1 expression in CC tissues was significantly associated with lower grade (χ2 = 3.592, P = 0.043), lower TNM stage (χ2 = 3.537, P = 0.048) and better overall survival (P=0.041). Jun N-terminal kinase 1 (JNK1) pathway and Mucin 2 (MUC2) were the potential downstream proteins of miR-613/ATOH1. miR-613 is an oncogene in CC and promotes the proliferation, invasion and migration of CC cells by targeting ATOH1 likely via activating JNK1 pathway and upregulating MUC2.
Collapse
Affiliation(s)
- Xuanxuan Yang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China
| | - Luo Zhang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China
| | - Xing Song
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China
| | - Wenting He
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China
| | - Dachuan Zhang
- Pathology Department, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China
| | - Qicheng Lu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China
| | - Jun Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China.
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou, 213003, China.
| |
Collapse
|
34
|
Cheng X, Yu D, Cheng G, Yung BC, Liu Y, Li H, Kang C, Fang X, Tian S, Zhou X, Liu Q, Lee RJ. T7 Peptide-Conjugated Lipid Nanoparticles for Dual Modulation of Bcl-2 and Akt-1 in Lung and Cervical Carcinomas. Mol Pharm 2018; 15:4722-4732. [PMID: 30138565 DOI: 10.1021/acs.molpharmaceut.8b00696] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Expression of Bcl-2 and Akt-1 has been associated with human cancer. G3139 and RX-0201, targeting Bcl-2 and Akt-1, respectively, are antisense oligonucleotides (ASOs) that have shown limited efficacy in clinical trials. Herein, we report a combination of newly designed ASOs based on these agents and was delivered by tumor cell-targeting lipid nanoparticles (LNPs). A "Gapmer" design strategy was applied to these ASOs with the addition of 2'-O-methyl modifications on the nucleotides at 5' and 3' ends. A dual-channel syringe pump-based system was developed for the synthesis of the LNPs. ASO-LNPs composed of DODMA, egg PC, cholesterol, T7-PEG-DSPE, and PEG-DMG at a molar ratio of 35:39.5:20:0.5:5 and carrying either individual ASOs or co-loaded ASO combinations (Co-ASOs) were synthesized and evaluated in both KB and A549 cancer cells and in an A549 murine xenograft model to determine their antitumor effects and biological activities. The ASO-LNPs exhibited excellent colloidal stability and high ASO encapsulation efficiency with relatively small mean particle sizes and moderately positive zeta potentials. Transferrin receptor-targeting T7-conjugated LNPs showed enhanced cellular uptake compared to nontargeted LNPs. In addition, both T7-conjugated Co-ASOs-LNPs and non-T7-conjugated Co-ASOs-LNPs at a molar ratio of (G3139-GAP to RX-0201-GAP at 1:2) showed efficient downregulation of both Bcl-2 and Akt-1 in both A549 and KB cells. Furthermore, T7-conjugated Co-ASOs-LNPs (Co-ASOs-LNPs) produced superior antitumor activity, prolonged the overall survival time, and demonstrated tumor targeting activity in an A549 xenograft model.
Collapse
Affiliation(s)
| | - Daorui Yu
- Department of Pharmacology, School of Basic Medicine and Life Science , Hainan Medical University , Haikou , China
| | - Guang Cheng
- State Key Laboratory of Long-Acting and Targeted Drug Delivery, Nanjing , China.,Luye Sike Pharma, Nanjing Hightech Industrial Development Zone, Nanjing , China
| | - Bryant C Yung
- The WhiteOak Group, LLC., Washington , D.C. 20006 , United States
| | | | | | - Chen Kang
- Department of Internal Medicine, Division of Cardiovascular Medicine, Carver College of Medicine , University of Iowa , Iowa City , Iowa 52242 , United States
| | - Xingyue Fang
- Department of Pharmacology, School of Basic Medicine and Life Science , Hainan Medical University , Haikou , China
| | - Shuhong Tian
- Department of Pharmacology, School of Basic Medicine and Life Science , Hainan Medical University , Haikou , China
| | - Xiaoju Zhou
- School of Pharmaceutical Science , Wuhan University , Wuhan 430071 , P.R. China
| | - Qibing Liu
- Department of Pharmacology, School of Basic Medicine and Life Science , Hainan Medical University , Haikou , China
| | | |
Collapse
|
35
|
Li J, Ghatak S, El Masry MS, Das A, Liu Y, Roy S, Lee RJ, Sen CK. Topical Lyophilized Targeted Lipid Nanoparticles in the Restoration of Skin Barrier Function following Burn Wound. Mol Ther 2018; 26:2178-2188. [PMID: 29802017 DOI: 10.1016/j.ymthe.2018.04.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 12/25/2022] Open
Abstract
Lyophilized keratinocyte-targeted nanocarriers (TLNκ) loaded with locked nucleic acid (LNA) modified anti-miR were developed for topical application to full thickness burn injury. TLNκ were designed to selectively deliver LNA-anti-miR-107 to keratinocytes using the peptide sequence ASKAIQVFLLAG. TLNκ employed DOTAP/DODAP combination pH-responsive lipid components to improve endosomal escape. To minimize interference of clearance by non-targeted cells, especially immune cells in the acute wound microenvironment, surface charge was neutralized. Lyophilization was performed to extend the shelf life of the lipid nanoparticles (LNPs). Encapsulation efficiency of anti-miR in lyophilized TLNκ was estimated to be 96.54%. Cargo stability of lyophilized TLNκ was tested. After 9 days of loading with anti-miR-210, TLNκ was effective in lowering abundance of the hypoxamiR miR-210 in keratinocytes challenged with hypoxia. Keratinocyte uptake of DiD-labeled TLNκ was selective and exceeded 90% within 4 hr. Topical application of hydrogel-dispersed lyophilized TLNκ encapsulating LNA anti-miR-107 twice a week significantly accelerated wound closure and restoration of skin barrier function. TLNκ/anti-miR-107 application depleted miR-107 and upregulated dicer expression, which accelerated differentiation of keratinocytes. Expression of junctional proteins such as claudin-1, loricrin, filaggrin, ZO-1, and ZO-2 were significantly upregulated following TLNκ/anti-miR-107 treatment. These LNPs are promising as topical therapeutic agents in the management of burn injury.
Collapse
Affiliation(s)
- Jilong Li
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Subhadip Ghatak
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Mohamed S El Masry
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Department of General Surgery (Plastic Surgery Unit), Zagazig University, 44519, Egypt
| | - Amitava Das
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Yang Liu
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Sashwati Roy
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Robert J Lee
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Chandan K Sen
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
36
|
Labatut AE, Mattheolabakis G. Non-viral based miR delivery and recent developments. Eur J Pharm Biopharm 2018; 128:82-90. [PMID: 29679644 DOI: 10.1016/j.ejpb.2018.04.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/28/2018] [Accepted: 04/18/2018] [Indexed: 12/18/2022]
Abstract
miRNAs are promising therapeutic targets or tools for the treatment of numerous diseases, with most prominently, cancer. The inherent capacity of these short nucleic acids to regulate multiple cancer-related pathways simultaneously has prompted strong research on understanding miR functions and their potential use for therapeutic purposes. A key determinant of miR therapeutics' potential for treatment is their delivery. Viral and non-viral vectors attempt to address the major limitations associated with miR delivery, but several hurdles have been identified. Here, we present an overview on the general limitations of miR delivery, and the delivery strategies exploited to overcome them. We provide an introduction on the advantages and disadvantages of viral and non-viral vectors, and we go into detail to analyze the most prominently used non-viral systems. We provide with an update on the most recent research on this topic and we describe the mechanism and limitations of the lipid-, polymer- and inorganic material- based miR delivery systems.
Collapse
Affiliation(s)
- Annalise Elizabeth Labatut
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, United States
| | - George Mattheolabakis
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, United States.
| |
Collapse
|
37
|
Li H, Liu Y, Chen L, Liu Q, Qi S, Cheng X, Lee YB, Ahn CH, Kim DJ, Lee RJ. Folate receptor-targeted lipid-albumin nanoparticles (F-LAN) for therapeutic delivery of an Akt1 antisense oligonucleotide. J Drug Target 2018; 26:466-473. [PMID: 29376449 DOI: 10.1080/1061186x.2018.1433678] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND RX-0201 is an antisense oligonucleotide (ASO) against Akt1 currently in clinical trial for metastatic renal cancer. PURPOSE To improve the delivery of RX-0201 using folate receptor-targeted lipid-albumin nanoparticles (F-LAN). METHODS F-LAN were synthesized with the composition of DOTAP/soyPC/TPGS/folate-PEG-DSPE (25:70:4:1 m/m), a cationic human serum albumin-pentaethylenehexamine (HSA-PEHA) conjugate and RX-0201. The nanoparticles were evaluated in KB human carcinoma cells in vitro and in a KB murine xenograft tumour model in vivo for pharmacokinetics and antitumor activities. RESULTS The F-LAN-RX-0201 had a mean particle size of 108.6 ± 5.8 nm, zeta potential of 10.5 ± 3.2 mV and ASO loading efficiency of 71.5 ± 4.5%. In KB cells, uptake and Akt1 inhibition by F-LAN-RX-0201 were greater than those of non-targeted LAN-RX-0201 and could be partially blocked by excess free folate. F-LAN-RX-0201 inhibited cell growth with an IC50 of 11.9 μM. In contrast, LAN-RX-0201 showed lower cytotoxicity with an IC50 of 32.0 μM. No significant cytotoxicity was observed with up to 250 µM of free RX-0201. Pharmacokinetic studies showed that F-LAN-RX-0201 had a longer terminal half-life than free RX-0201 (442 vs. 219 min). In a KB xenograft tumour model, F-LAN-RX-0201 exhibited greater tumour inhibition than LAN-RX-0201 at 16 mg/kg. Moreover, F-LAN-RX-0201 at 16 mg/kg showed comparable tumour inhibition compared to free RX-0201 at a much higher dose of 90 mg/kg. CONCLUSIONS F-LAN-RX-0201 showed promise as a therapeutic agent for tumours with elevated folate-receptor expression.
Collapse
Affiliation(s)
- Hong Li
- a Division of Pharmaceutics , College of Pharmacy, The Ohio State University , Columbus , OH , USA
| | - Yang Liu
- a Division of Pharmaceutics , College of Pharmacy, The Ohio State University , Columbus , OH , USA
| | - Lihua Chen
- a Division of Pharmaceutics , College of Pharmacy, The Ohio State University , Columbus , OH , USA
| | - Qibing Liu
- a Division of Pharmaceutics , College of Pharmacy, The Ohio State University , Columbus , OH , USA
| | - Shanshan Qi
- a Division of Pharmaceutics , College of Pharmacy, The Ohio State University , Columbus , OH , USA
| | - Xinwei Cheng
- a Division of Pharmaceutics , College of Pharmacy, The Ohio State University , Columbus , OH , USA
| | - Young B Lee
- b Rexahn Pharmaceuticals, Inc. , Rockville , MD , USA
| | - Chang-Ho Ahn
- b Rexahn Pharmaceuticals, Inc. , Rockville , MD , USA
| | | | - Robert J Lee
- a Division of Pharmaceutics , College of Pharmacy, The Ohio State University , Columbus , OH , USA
| |
Collapse
|
38
|
Tian Y, Zhang C, Guo M. Comparative study on alkaloids and their anti-proliferative activities from three Zanthoxylum species. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:460. [PMID: 28899423 PMCID: PMC5596839 DOI: 10.1186/s12906-017-1966-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/01/2017] [Indexed: 11/25/2022]
Abstract
Background Alkaloids have been considered as the most promising bioactive ingredients in plant species from the genus Zanthoxylum. This study reports on the compositions and contents of the Zanthoxylum alkaloids (ZAs) from three Zanthoxylum species, and their potential anti-proliferation activities. Methods An HPLC-UV/ESI-MS/MS method was established and employed to analyze the alkaloids in different Zanthoxylum extracts. The common and unique peaks and their relative contents were summarized and compared to evaluate the similarity and dissimilarity of the three Zanthoxylum species. Meanwhile, inhibitory activity tests to four carcinoma cell lines, i.e., stomach tumor cells (SGC-7901), cervical tumor cells (Hela), colon tumor cells (HT-29) and Hepatic tumor cells (Hep G2), were carried out in vitro to evaluate the bioactivities of the ZAs. Results Seventy peaks were detected in the crude total alkaloid samples, and 58 of them were identified. As a result, 13 common peaks were found in the extracts of all the three Zanthoxylum species, while some unique peaks were also observed in specific species, with 17 peaks in Z. simulans, 15 peaks in Z. ailanthoides and 11 peaks in Z. chalybeum, respectively. The comparison of the composition and relative contents indicated that alkaloids of benzophenanthridine type commonly present in all the three Zanthoxylum species with high relative contents among the others, which are 60.52% in Z. ailanthoides, 30.52% in Z. simulans and 13.84% in Z. chalybeum, respectively. In terms of activity test, Most of the crude alkaloids extracts showed remarkable inhibitory activities against various tumor cells, and the inhibitory rates ranged from 60.71 to 93.63% at a concentration of 200 μg/mL. However, SGC-7901 cells seemed to be more sensitive to the ZAs than the other three cancer cells. Conclusion The alkaloid profiles detected in this work revealed significant differences in both structures and contents among Zanthoxylum species. The inhibitory rates for different cancer cells in this study indicated that the potential anti-cancer activity should be attributed to quaternary alkaloids in these three species, which will provide great guidance for further exploring this traditional medicinal resource as new healthcare products.
Collapse
|
39
|
Sun Y, Kang C, Liu F, Zhou Y, Luo L, Qiao H. RGD Peptide-Based Target Drug Delivery of Doxorubicin Nanomedicine. Drug Dev Res 2017; 78:283-291. [PMID: 28815721 DOI: 10.1002/ddr.21399] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Preclinical Research Doxorubicin (DOX) is commonly used for the treatment of breast cancer and lymphoma. However, its clinical use has been severely limited due to cardiotoxicity, requiring the development of safer and more efficient pharmaceutical formulations of DOX. Advances in nanotechnology have provided new ways to administer chemotherapeutic drugs like DOX are conveyed into the body and to tumor sites. These Nanotechnology approaches have aided in the selective accumulation of DOX into tumor sites via the enhanced permeability and retention. However, the absence of active targeting ligands still hinders the effective delivery of DOX. Among all active targeting ligands developed to date, RGD peptide (Arginylglycylaspartic acid) occupies a unique position owing to its inherent safety, biocompatibility, and targeting ability. Accordingly, modification of DOX with RGD ligand is anticipated to improve transport of DOX into tumor cells. In this review, we discuss using RGD peptide for improving the therapeutic efficacy of DOX nanomedicine. Drug Dev Res 78 : 283-291, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, Sacramento, California, 95758
| | - Chen Kang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, 52242
| | - Fei Liu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - You Zhou
- College of Biotechnology, Southwest University, Chongqing, 400715, China
| | - Lei Luo
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Hongzhi Qiao
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
40
|
MiR-21 is required for anti-tumor immune response in mice: an implication for its bi-directional roles. Oncogene 2017; 36:4212-4223. [PMID: 28346427 DOI: 10.1038/onc.2017.62] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/29/2016] [Accepted: 02/08/2017] [Indexed: 12/17/2022]
Abstract
Here we show that miR-21, a microRNA known for its oncogenic activity, is also essential for mediating immune responses against tumor. Knockout of miR-21 in mice slowed the proliferation of both CD4+ and CD8+ cells, reduced their cytokine production and accelerated the grafted tumor growth. Further investigations indicated that miR-21 could activate CD4+ and CD8+ T cells via the PTEN/Akt pathway in response to stimulations. Taken together, these data suggest the key functions of miR-21 in mediating anti-tumor immune response and thereby uncover a bi-directional role of this traditionally known 'oncomiR' in tumorigenesis. Our study may provide new insights for the design of cancer therapies targeting microRNAs, with an emphasis on the dynamic and possibly unexpected role of these molecules.
Collapse
|
41
|
Lai X, Friedman A. Exosomal miRs in Lung Cancer: A Mathematical Model. PLoS One 2016; 11:e0167706. [PMID: 28002496 PMCID: PMC5176278 DOI: 10.1371/journal.pone.0167706] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 11/18/2016] [Indexed: 01/11/2023] Open
Abstract
Lung cancer, primarily non-small-cell lung cancer (NSCLC), is the leading cause of cancer deaths in the United States and worldwide. While early detection significantly improves five-year survival, there are no reliable diagnostic tools for early detection. Several exosomal microRNAs (miRs) are overexpressed in NSCLC, and have been suggested as potential biomarkers for early detection. The present paper develops a mathematical model for early stage of NSCLC with emphasis on the role of the three highest overexpressed miRs, namely miR-21, miR-205 and miR-155. Simulations of the model provide quantitative relationships between the tumor volume and the total mass of each of the above miRs in the tumor. Because of the positive correlation between these miRs in the tumor tissue and in the blood, the results of the paper may be viewed as a first step toward establishing a combination of miRs 21, 205, 155 and possibly other miRs as serum biomarkers for early detection of NSCLC.
Collapse
Affiliation(s)
- Xiulan Lai
- Institute for Mathematical Sciences, Renmin University of China, Beijing, P. R. China
| | - Avner Friedman
- Mathematical Bioscience Institute & Department of Mathematics, Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
42
|
Cheng X, Liu Q, Li H, Kang C, Liu Y, Guo T, Shang K, Yan C, Cheng G, Lee RJ. Lipid Nanoparticles Loaded with an Antisense Oligonucleotide Gapmer Against Bcl-2 for Treatment of Lung Cancer. Pharm Res 2016; 34:310-320. [PMID: 27896589 DOI: 10.1007/s11095-016-2063-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/01/2016] [Indexed: 12/19/2022]
Abstract
PURPOSE Bcl-2 is an anti-apoptotic gene that is frequently overexpressed in human cancers. G3139 is an antisense oligonucleotide against bcl-2 that has shown limited efficacy in clinical trials. Here, we report the synthesis of a new antisense oligonucleotide containing additional chemical modifications and its delivery using nanoparticles. METHODS An oligonucleotide G3139-GAP was synthesized, which has 2'-O-methyl nucleotides at the 5' and 3' ends based on a "gapmer" design. Furthermore, G3139-GAP was incorporated into lipid nanoparticles (LNPs) composed of DOTAP/egg PC/cholesterol/Tween 80. The LNP-loaded G3139-GAP was evaluated in A549 lung cancer cells both in vitro and in a murine xenograft model for biological activity and therapeutic efficacy. RESULTS The LNPs showed excellent colloidal and serum stability, and high encapsulation efficiency for G3139-GAP. They have a mean particle diameter and zeta potential of 134 nm and 9.59 mV, respectively. G3139-GAP-LNPs efficiently downregulated bcl-2 expression in A549 cells, as shown by 40% and 83% reduction in mRNA and protein levels, respectively. Furthermore, G3139-GAP-LNPs were shown to inhibit tumor growth, prolong survival, and downregulate tumor bcl-2 expression in an A549 murine xenograft tumor model. These data indicate that G3139-GAP-LNPs have excellent anti-tumor efficacy and warrant further evaluation.
Collapse
Affiliation(s)
- Xinwei Cheng
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Qibing Liu
- Department of Pharmacology, Hainan Medical University, Xueyuan Road, Haikou, 571199, Hainan, China
| | - Hong Li
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, 500 West 12th Ave., Columbus, Ohio, 43210, USA
| | - Chen Kang
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Yang Liu
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, 500 West 12th Ave., Columbus, Ohio, 43210, USA
| | - Tianqi Guo
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, 500 West 12th Ave., Columbus, Ohio, 43210, USA
| | - Ke Shang
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, 500 West 12th Ave., Columbus, Ohio, 43210, USA
| | - Chengyun Yan
- First Affiliated Hospital of Jiamusi University, Jiamusi University, Jiamusi, 154007, Heilongjiang, China
| | - Guang Cheng
- State Key Laboratory of Long-Acting and Targeted Drug Delivery, Nanjing, 210061, Jiangsu, China. .,Nanjing Hightech Industrial Development Zone, 28 Gaoxin Road, Nanjing, 210061, Jiangsu, China.
| | - Robert J Lee
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, 43210, USA. .,Division of Pharmaceutics, College of Pharmacy, The Ohio State University, 500 West 12th Ave., Columbus, Ohio, 43210, USA.
| |
Collapse
|
43
|
Liu F, Sun Y, Kang C, Zhu H. Pegylated Drug Delivery Systems: From Design to Biomedical Applications. ACTA ACUST UNITED AC 2016. [DOI: 10.1142/s1793984416420022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pegylation, as a simple procedure to attach hydrophilic polyethylene glycol (PEG) onto therapeutic molecule or drug carriers has been utilized widely to deliver small molecules, proteins and peptides. It was first reported in 1970s by Dr. Frank Davis of Rutgers University and Dr. Abuchowsky in the studies of PEG modified albumin and catalase. The significance of this method at that time was able to successfully modify the enzyme with better hydrophilicity but also keep the enzymatic activity. The employment of PEG has provided superior stability of drug delivery systems (DDS) and enhanced the circulation time in vivo. Simple conjugation of PEG chains with various molecular weights enables the possibility to regulate the properties of desired DDS and led to important contribution in targeting therapy and diagnosis. Pegylation has been reported to be able to protect peptides by shielding antigenic epitopes from reticuloendothelial (RES) clearance and avoid enzymes being recognized by immune system and avoid early degradation. In addition, utilization of PEG in DDS are reported with enhanced delivery efficiency, prolonged circulation time and improved stability, especially active enzymes and peptides drug delivery. In this paper, we will conclude current studies about Pegylated DDS and their biomedical applications from both in vitro and in vivo studies.
Collapse
Affiliation(s)
- Fei Liu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35209, USA
| | - Yuan Sun
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Chen Kang
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, IA 52242, USA
| | - Hongyan Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, P. R. China
| |
Collapse
|
44
|
Sun Y, Kang C, Yao Z, Liu F, Zhou Y. Peptide-Based Ligand for Active Delivery of Liposomal Doxorubicin. ACTA ACUST UNITED AC 2016. [DOI: 10.1142/s1793984416420046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Doxorubicin (DOX) has been extensively used in the clinic to treat malignant tumors such as leukemias and Hodgkin’s lymphoma. However, the severe cardiotoxicity associated with the use of DOX requests the development of alternative and efficient pharmaceutical formulations. The PEGylated liposome of DOX can significantly reduce the cardiotoxicity but still lacks the active targeting towards cancer cells. Modification of liposomal DOX with active ligands would then be a rational approach to enhance the transportation of the toxin into tumor cells. Currently used targeting ligands include antibodies, proteins, small molecules, and peptides. By virtue of the advantages such as easy preparation, lower cost, and elevated resistance to enzymatic degradation, peptides are attracting a significant amount of interest as active targeting ligands for pharmaceutics. In this paper, we will briefly discuss the application of peptide ligands for the improvement of the therapeutic efficacy of liposomal DOX.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Chen Kang
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhili Yao
- Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Fei Liu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - You Zhou
- College of Biotechnology, Southwest University, Chongqing, 400715, P. R. China
| |
Collapse
|
45
|
Sun Y, Kang C, Liu F, Song L. Delivery of Antipsychotics with Nanoparticles. Drug Dev Res 2016; 77:393-399. [PMID: 27546713 DOI: 10.1002/ddr.21331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 06/16/2016] [Indexed: 01/05/2023]
Abstract
Preclinical Research Psychosis remains one of the most challenging health problems for society, affecting hundreds of millions of people worldwide. Although current antipsychotics can alleviate the symptoms of psychosis, they are still far away from being perfect, often causing significant and even fatal side effects such as involuntary movement disorders and metabolic syndrome. With the lack of precise knowledge of the underlying mechanisms of psychosis, a rational approach to improve the efficiency of current antipsychotics is by nanoparticle-based administration. Nanoparticles with the size of 1-500 nm can be used in drug formulations to pass through many biological barriers including the blood-brain barrier, which makes them excellent candidates for the delivery of antipsychotics. Besides that, nanoparticles loaded with antipsychotics can solve the common aqueous solubility issues for most brain targeting drugs, and enable a slow-release profile for the encapsulated drugs. This research overview provides a brief summary and discussion of the progress and development in the delivery of antipsychotics with nanoparticle formulations over the past five years (2011-2016). Drug Dev Res 77 : 393-399, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio, 43210
| | - Chen Kang
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, 43210
| | - Fei Liu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Lei Song
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida, 33136
| |
Collapse
|
46
|
Hu GF, Quan RF, Chen YM, Bi DW, Jiang XS, Li XF, Li JY. Fabrication, characterization, bioactivity, and biocompatibility of novel mesoporous calcium silicate/polyetheretherketone composites. RSC Adv 2016. [DOI: 10.1039/c6ra07272k] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Composite consisting of polyetheretherketone and mesoporous calcium silicate were fabricated. The composite with improved hydrophilicity, bioactivity and biocompatibility might be a great candidate for bone tissue regeneration.
Collapse
Affiliation(s)
- G. F. Hu
- The First People's Hospital of Xiaoshang
- Hangzhou 311200
- China
| | - R. F. Quan
- Xiaoshang Hospital of Traditional Chinese Medicine
- Hangzhou 311200
- China
| | - Y. M. Chen
- The First People's Hospital of Xiaoshang
- Hangzhou 311200
- China
| | - D. W. Bi
- The First People's Hospital of Xiaoshang
- Hangzhou 311200
- China
| | - X. S. Jiang
- Orthopedics Department
- Huzhou Central Hospital
- Huzhou 313000
- China
| | - X. F. Li
- Orthopedics Department
- Huzhou Central Hospital
- Huzhou 313000
- China
| | - J. Y. Li
- Orthopedics Department
- Huzhou Central Hospital
- Huzhou 313000
- China
| |
Collapse
|