1
|
Liao S, Li S, Liu Z, Lu W, He Y, Xia K, Wang Y, Zhao Z, Lin Y. A bioswitchable siRNA delivery system: RNAi therapy based on tetrahedral framework nucleic acids for bone defect repair. NANOSCALE 2024; 16:21531-21544. [PMID: 39480485 DOI: 10.1039/d4nr04105d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Craniofacial bone defects, caused by trauma, congenital abnormalities, or various diseases, present a significant challenge in regenerative medicine. One approach to addressing this problem is the use of RNA interference (RNAi) technology with small interfering RNA (siRNA). CKIP-1 is a negative regulatory molecule for bone formation. However, direct applications of CKIP-1 siRNA for bone defects are still limited. The instability and poor cellular uptake ability of CKIP-1 siRNA restrict its clinical applications. A new drug delivery system is critically needed to enhance the effectiveness and potential applications of CKIP-1 siRNA. Tetrahedral framework nucleic acid (tFNA) is a promising drug delivery system due to its stability and transport abilities. In this study, we developed a bioswitchable siRNA delivery system (BiRDS) based on tFNA to carry CKIP-1 siRNA and examined its effect on bone defect repair. siRNA was successfully loaded into the tFNA core, forming BiRDS, which improved siRNA stability and cellular uptake. After entering cells, BiRDS exposed siRNA, enhancing CKIP-1 silencing efficiency. This system significantly promoted osteogenic differentiation and bone regeneration in rat mandibular bone defects, offering a new strategy for bone regeneration therapy.
Collapse
Affiliation(s)
- Shengnan Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Songhang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Zhiqiang Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Weitong Lu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yutian He
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Kai Xia
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yigan Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
2
|
Smail SW, Hirmiz SM, Ahmed AA, Albarzinji N, Awla HK, Amin K, Janson C. Decoding the intricacies: a comprehensive analysis of microRNAs in the pathogenesis, diagnosis, prognosis and therapeutic strategies for COVID-19. Front Med (Lausanne) 2024; 11:1430974. [PMID: 39434774 PMCID: PMC11492531 DOI: 10.3389/fmed.2024.1430974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
The pandemic of coronavirus disease-19 (COVID-19), provoked by the appearance of a novel coronavirus named severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), required a worldwide healthcare emergency. This has elicited an immediate need for accelerated research into its mechanisms of disease, criteria for diagnosis, methods for forecasting outcomes, and treatment approaches. microRNAs (miRNAs), are diminutive RNA molecules, that are non-coding and participate in gene expression regulation post-transcriptionally, having an important participation in regulating immune processes. miRNAs have granted substantial interest in their impact on viral replication, cell proliferation, and modulation of how the host's immune system responds. This narrative review delves into host miRNAs' multifaceted roles within the COVID-19 context, highlighting their involvement in disease progression, diagnostics, and prognostics aspects, given their stability in biological fluids and varied expression profiles when responding to an infection. Additionally, we discuss complicated interactions between SARS-CoV-2 and host cellular machinery facilitated by host miRNAs revealing how dysregulation of host miRNA expression profiles advances viral replication, immune evasion, and inflammatory responses. Furthermore, it investigates the potential of host miRNAs as therapeutic agents, whether synthetic or naturally occurring, which could be harnessed to either mitigate harmful inflammation or enhance antiviral responses. However, searching more deeply is needed to clarify how host's miRNAs are involved in pathogenesis of COVID-19, its diagnosis processes, prognostic assessments, and treatment approaches for patients.
Collapse
Affiliation(s)
- Shukur Wasman Smail
- College of Pharmacy, Cihan University-Erbil, Kurdistan Region, Erbil, Iraq
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Sarah Mousa Hirmiz
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Akhter Ahmed Ahmed
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Niaz Albarzinji
- Department of Medicine, Hawler Medical University, Erbil, Iraq
| | - Harem Khdir Awla
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Kawa Amin
- College of Medicine, University of Sulaimani, Sulaymaniyah, Iraq
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Christer Janson
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| |
Collapse
|
3
|
Wang P, Zhou Y, Wang J, Zhou Y, Zhang X, Liu Y, Li A, He Y, Chen S, Qian A, Wang X, Nie Y, Fan D, Cao T, Lu Y, Zhao X. miR-107 reverses the multidrug resistance of gastric cancer by targeting the CGA/EGFR/GATA2 positive feedback circuit. J Biol Chem 2024; 300:107522. [PMID: 38960034 PMCID: PMC11345541 DOI: 10.1016/j.jbc.2024.107522] [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: 12/27/2023] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024] Open
Abstract
Chemotherapy is still the main therapeutic strategy for gastric cancer (GC). However, most patients eventually acquire multidrug resistance (MDR). Hyperactivation of the EGFR signaling pathway contributes to MDR by promoting cancer cell proliferation and inhibiting apoptosis. We previously identified the secreted protein CGA as a novel ligand of EGFR and revealed a CGA/EGFR/GATA2 positive feedback circuit that confers MDR in GC. Herein, we outline a microRNA-based treatment approach for MDR reversal that targets both CGA and GATA2. We observed increased expression of CGA and GATA2 and increased activation of EGFR in GC samples. Bioinformatic analysis revealed that miR-107 could simultaneously target CGA and GATA2, and the low expression of miR-107 was correlated with poor prognosis in GC patients. The direct interactions between miR-107 and CGA or GATA2 were validated by luciferase reporter assays and Western blot analysis. Overexpression of miR-107 in MDR GC cells increased their susceptibility to chemotherapeutic agents, including fluorouracil, adriamycin, and vincristine, in vitro. Notably, intratumor injection of the miR-107 prodrug enhanced MDR xenograft sensitivity to chemotherapies in vivo. Molecularly, targeting CGA and GATA2 with miR-107 inhibited EGFR downstream signaling, as evidenced by the reduced phosphorylation of ERK and AKT. These results suggest that miR-107 may contribute to the development of a promising therapeutic approach for the treatment of MDR in GC.
Collapse
Affiliation(s)
- Pei Wang
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China; Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yelu Zhou
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Juan Wang
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yun Zhou
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaohui Zhang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yanxing Liu
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ang Li
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yangsong He
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shuyi Chen
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China; School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Airong Qian
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Xin Wang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yongzhan Nie
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Daiming Fan
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tianyu Cao
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Yuanyuan Lu
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Xiaodi Zhao
- Xijing Hospital of Digestive Diseases, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
4
|
Naimi N, Seyedmirzaei H, Hassannejad Z, Soltani Khaboushan A. Advanced nanoparticle strategies for optimizing RNA therapeutic delivery in neurodegenerative disorders. Biomed Pharmacother 2024; 175:116691. [PMID: 38713941 DOI: 10.1016/j.biopha.2024.116691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/09/2024] Open
Abstract
Neurodegenerative diseases affect many people worldwide, and as the population ages, the incidence of these conditions increases. Alzheimer's disease (AD) and Parkinson's disease (PD) are the most prevalent neurodegenerative disorders worldwide. Different medicines are being used to control symptoms related to these conditions, but no treatment has yet been approved. Both genetic and environmental factors are involved in disease pathogenesis, and research on the pathophysiological pathways is still ongoing. The role of subcellular pathways and dysregulation in RNA pathways has been highlighted in pathophysiological studies, and treatment strategies focused on these pathways can be a promising approach. Many experiments have been conducted on delivering RNA cargo to the CNS to modulate various pathways involved. Yet another challenge to be faced is the effective transport of desired molecules to targets, which can be greatly hindered by distinct barriers limiting transport to the CNS, most noticeably the blood-brain barrier (BBB). Nanotechnology and the use of different nano-carriers for the delivery of nucleotides, peptides, proteins, and drug molecules are currently of great interest as these carriers help with better delivery and protection and, as a result, improve the effectiveness of the cargo. Nanocarriers can protect susceptible RNA molecules from possible degradation or destruction and improve their ability to reach the brain by enhancing BBB penetration. Different mechanisms for this process have been hypothesized. This review will go through the therapeutic application of RNA molecules in the treatment of AD and PD and the role of nanocarriers in overcoming delivery challenges and enhancing efficacy.
Collapse
Affiliation(s)
- Narges Naimi
- Departement of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Homa Seyedmirzaei
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran.
| | - Alireza Soltani Khaboushan
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran; Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Tao T, Rehman SU, Xu S, Zhang J, Xia H, Guo Z, Li Z, Ma K, Wang J. A biomimetic camouflaged metal organic framework for enhanced siRNA delivery in the tumor environment. J Mater Chem B 2024; 12:4080-4096. [PMID: 38577851 DOI: 10.1039/d3tb02827e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Gene silencing through RNA interference (RNAi), particularly using small double-stranded RNA (siRNA), has been identified as a potent strategy for targeted cancer treatment. Yet, its application faces challenges such as nuclease degradation, inefficient cellular uptake, endosomal entrapment, off-target effects, and immune responses, which have hindered its effective delivery. In the past few years, these challenges have been addressed significantly by using camouflaged metal-organic framework (MOF) nanocarriers. These nanocarriers protect siRNA from degradation, enhance cellular uptake, and reduce unintended side effects by effectively targeting desired cells while evading immune detection. By combining the properties of biomimetic membranes and MOFs, these nanocarriers offer superior benefits such as extended circulation times, enhanced stability, and reduced immune responses. Moreover, through ligand-receptor interactions, biomimetic membrane-coated MOFs achieve homologous targeting, minimizing off-target adverse effects. The MOFs, acting as the core, efficiently encapsulate and protect siRNA molecules, while the biomimetic membrane-coated surface provides homologous targeting, further increasing the precision of siRNA delivery to cancer cells. In particular, the biomimetic membranes help to shield the MOFs from the immune system, avoiding unwanted immune responses and improving their biocompatibility. The combination of siRNA with innovative nanocarriers, such as camouflaged-MOFs, presents a significant advancement in cancer therapy. The ability to deliver siRNA with precision and effectiveness using these camouflaged nanocarriers holds great promise for achieving more personalized and efficient cancer treatments in the future. This review article discusses the significant progress made in the development of siRNA therapeutics for cancer, focusing on their effective delivery through novel nanocarriers, with a particular emphasis on the role of metal-organic frameworks (MOFs) as camouflaged nanocarriers.
Collapse
Affiliation(s)
- Tongxiang Tao
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- University of Science and Technology of China, Hefei 230036, Anhui, P. R. China
| | - Sajid Ur Rehman
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
| | - Shuai Xu
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Jing Zhang
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Haining Xia
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Zeyong Guo
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Zehua Li
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- Hefei Cancer Hospital, Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, Anhui, P. R. China
| | - Kun Ma
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
| | - Junfeng Wang
- CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, P. R. China.
- University of Science and Technology of China, Hefei 230036, Anhui, P. R. China
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, Anhui, P. R. China
| |
Collapse
|
6
|
Drago SE, Cabibbo M, Craparo EF, Cavallaro G. TAT decorated siRNA polyplexes for inhalation delivery in anti-asthma therapy. Eur J Pharm Sci 2023; 190:106580. [PMID: 37717668 DOI: 10.1016/j.ejps.2023.106580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/20/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023]
Abstract
In this work, a novel protonable copolymer was designed to deliver siRNA through the inhalation route, as an innovative formulation for the management of asthma. This polycation was synthesized by derivatization of α,β-poly(N-2-hydroxyethyl)D,L-aspartamide (PHEA) first with 1,2-Bis(3-aminopropylamino)ethane (bAPAE) and then with a proper amount of maleimide terminated poly(ethylene glycol) (PEG-MLB), with the aim to increase the superficial hydrophilicity of the system, allowing the diffusion trough the mucus layer. Once the complexation ability of the copolymer has been evaluated, obtaining nanosized polyplexes, polyplexes were functionalized on the surface with a thiolated TAT peptide, a cell-penetrating peptide (CPP), exploiting a thiol-ene reaction. TAT decorated polyplexes result to be highly cytocompatible and able to retain the siRNA with a suitable complexation weight ratio during the diffusion process through the mucus. Despite polyplexes establish weak bonds with the mucin chains, these can diffuse efficiently through the mucin layer and therefore potentially able to reach the bronchial epithelium. Furthermore, through cellular uptake studies, it was possible to observe how the obtained polyplexes penetrate effectively in the cytoplasm of bronchial epithelial cells, where they can reduce IL-8 gene expression, after LPS exposure. In the end, in order to obtain a formulation administrable as an inhalable dry powder, polyplexes were encapsulated in mannitol-based microparticles, by spray freeze drying, obtaining highly porous particles with proper technological characteristics that make them potentially administrable by inhalation route.
Collapse
Affiliation(s)
- Salvatore Emanuele Drago
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy
| | - Marta Cabibbo
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy
| | - Emanuela Fabiola Craparo
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy
| | - Gennara Cavallaro
- Lab of Biocompatible Polymers, Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, Palermo 90123, Italy; Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM) of Palermo, Palermo, Italy; Advanced Technology and Network Center (ATeN Center), Università di Palermo, Palermo 90133, Italy.
| |
Collapse
|
7
|
Won Lee J, Kyu Shim M, Kim H, Jang H, Lee Y, Hwa Kim S. RNAi therapies: Expanding applications for extrahepatic diseases and overcoming delivery challenges. Adv Drug Deliv Rev 2023; 201:115073. [PMID: 37657644 DOI: 10.1016/j.addr.2023.115073] [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: 04/27/2023] [Revised: 07/31/2023] [Accepted: 08/20/2023] [Indexed: 09/03/2023]
Abstract
The era of RNA medicine has become a reality with the success of messenger RNA (mRNA) vaccines against COVID-19 and the approval of several RNA interference (RNAi) agents in recent years. Particularly, therapeutics based on RNAi offer the promise of targeting intractable and previously undruggable disease genes. Recent advances have focused in developing delivery systems to enhance the poor cellular uptake and insufficient pharmacokinetic properties of RNAi therapeutics and thereby improve its efficacy and safety. However, such approach has been mainly achieved via lipid nanoparticles (LNPs) or chemical conjugation with N-Acetylgalactosamine (GalNAc), thus current RNAi therapy has been limited to liver diseases, most likely to encounter liver-targeting limitations. Hence, there is a huge unmet medical need for intense evolution of RNAi therapeutics delivery systems to target extrahepatic tissues and ultimately extend their indications for treating various intractable diseases. In this review, challenges of delivering RNAi therapeutics to tumors and major organs are discussed, as well as their transition to clinical trials. This review also highlights innovative and promising preclinical RNAi-based delivery platforms for the treatment of extrahepatic diseases.
Collapse
Affiliation(s)
- Jong Won Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Man Kyu Shim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hyosuk Kim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hochung Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Yuhan Lee
- Department of Anesthesiology, Perioperative, and Pain Medicine, Center for Accelerated Medical Innovation & Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Sun Hwa Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| |
Collapse
|
8
|
Ashique S, Garg A, Hussain A, Farid A, Kumar P, Taghizadeh‐Hesary F. Nanodelivery systems: An efficient and target-specific approach for drug-resistant cancers. Cancer Med 2023; 12:18797-18825. [PMID: 37668041 PMCID: PMC10557914 DOI: 10.1002/cam4.6502] [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: 02/01/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Cancer treatment is still a global health challenge. Nowadays, chemotherapy is widely applied for treating cancer and reducing its burden. However, its application might be in accordance with various adverse effects by exposing the healthy tissues and multidrug resistance (MDR), leading to disease relapse or metastasis. In addition, due to tumor heterogeneity and the varied pharmacokinetic features of prescribed drugs, combination therapy has only shown modestly improved results in MDR malignancies. Nanotechnology has been explored as a potential tool for cancer treatment, due to the efficiency of nanoparticles to function as a vehicle for drug delivery. METHODS With this viewpoint, functionalized nanosystems have been investigated as a potential strategy to overcome drug resistance. RESULTS This approach aims to improve the efficacy of anticancer medicines while decreasing their associated side effects through a range of mechanisms, such as bypassing drug efflux, controlling drug release, and disrupting metabolism. This review discusses the MDR mechanisms contributing to therapeutic failure, the most cutting-edge approaches used in nanomedicine to create and assess nanocarriers, and designed nanomedicine to counteract MDR with emphasis on recent developments, their potential, and limitations. CONCLUSIONS Studies have shown that nanoparticle-mediated drug delivery confers distinct benefits over traditional pharmaceuticals, including improved biocompatibility, stability, permeability, retention effect, and targeting capabilities.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of PharmaceuticsPandaveswar School of PharmacyPandaveswarIndia
| | - Ashish Garg
- Guru Ramdas Khalsa Institute of Science and Technology, PharmacyJabalpurIndia
| | - Afzal Hussain
- Department of Pharmaceutics, College of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Arshad Farid
- Gomal Center of Biochemistry and BiotechnologyGomal UniversityDera Ismail KhanPakistan
| | - Prashant Kumar
- Teerthanker Mahaveer College of PharmacyTeerthanker Mahaveer UniversityMoradabadIndia
- Department of Pharmaceutics, Amity Institute of PharmacyAmity University Madhya Pradesh (AUMP)GwaliorIndia
| | - Farzad Taghizadeh‐Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of MedicineIran University of Medical SciencesTehranIran
- Clinical Oncology DepartmentIran University of Medical SciencesTehranIran
| |
Collapse
|
9
|
Vora LK, Gholap AD, Jetha K, Thakur RRS, Solanki HK, Chavda VP. Artificial Intelligence in Pharmaceutical Technology and Drug Delivery Design. Pharmaceutics 2023; 15:1916. [PMID: 37514102 PMCID: PMC10385763 DOI: 10.3390/pharmaceutics15071916] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/28/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Artificial intelligence (AI) has emerged as a powerful tool that harnesses anthropomorphic knowledge and provides expedited solutions to complex challenges. Remarkable advancements in AI technology and machine learning present a transformative opportunity in the drug discovery, formulation, and testing of pharmaceutical dosage forms. By utilizing AI algorithms that analyze extensive biological data, including genomics and proteomics, researchers can identify disease-associated targets and predict their interactions with potential drug candidates. This enables a more efficient and targeted approach to drug discovery, thereby increasing the likelihood of successful drug approvals. Furthermore, AI can contribute to reducing development costs by optimizing research and development processes. Machine learning algorithms assist in experimental design and can predict the pharmacokinetics and toxicity of drug candidates. This capability enables the prioritization and optimization of lead compounds, reducing the need for extensive and costly animal testing. Personalized medicine approaches can be facilitated through AI algorithms that analyze real-world patient data, leading to more effective treatment outcomes and improved patient adherence. This comprehensive review explores the wide-ranging applications of AI in drug discovery, drug delivery dosage form designs, process optimization, testing, and pharmacokinetics/pharmacodynamics (PK/PD) studies. This review provides an overview of various AI-based approaches utilized in pharmaceutical technology, highlighting their benefits and drawbacks. Nevertheless, the continued investment in and exploration of AI in the pharmaceutical industry offer exciting prospects for enhancing drug development processes and patient care.
Collapse
Affiliation(s)
- Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Keshava Jetha
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
- Ph.D. Section, Gujarat Technological University, Ahmedabad 382424, Gujarat, India
| | | | - Hetvi K Solanki
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
10
|
Danckwardt S, Trégouët DA, Castoldi E. Post-transcriptional control of haemostatic genes: mechanisms and emerging therapeutic concepts in thrombo-inflammatory disorders. Cardiovasc Res 2023; 119:1624-1640. [PMID: 36943786 PMCID: PMC10325701 DOI: 10.1093/cvr/cvad046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 03/23/2023] Open
Abstract
The haemostatic system is pivotal to maintaining vascular integrity. Multiple components involved in blood coagulation have central functions in inflammation and immunity. A derailed haemostasis is common in prevalent pathologies such as sepsis, cardiovascular disorders, and lately, COVID-19. Physiological mechanisms limit the deleterious consequences of a hyperactivated haemostatic system through adaptive changes in gene expression. While this is mainly regulated at the level of transcription, co- and posttranscriptional mechanisms are increasingly perceived as central hubs governing multiple facets of the haemostatic system. This layer of regulation modulates the biogenesis of haemostatic components, for example in situations of increased turnover and demand. However, they can also be 'hijacked' in disease processes, thereby perpetuating and even causally entertaining associated pathologies. This review summarizes examples and emerging concepts that illustrate the importance of posttranscriptional mechanisms in haemostatic control and crosstalk with the immune system. It also discusses how such regulatory principles can be used to usher in new therapeutic concepts to combat global medical threats such as sepsis or cardiovascular disorders.
Collapse
Affiliation(s)
- Sven Danckwardt
- Centre for Thrombosis and Hemostasis (CTH), University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- German Centre for Cardiovascular Research (DZHK),
Berlin, Germany
- Posttranscriptional Gene Regulation, University Medical Centre
Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University
Medical Centre Mainz, Langenbeckstr. 1, 55131
Mainz, Germany
- Center for Healthy Aging (CHA), Mainz,
Germany
| | - David-Alexandre Trégouët
- INSERM, Bordeaux Population Health Research Center, UMR 1219, Department of
Molecular Epidemiology of Vascular and Brain Disorders (ELEANOR), University of
Bordeaux, Bordeaux, France
| | - Elisabetta Castoldi
- Department of Biochemistry, Cardiovascular Research Institute Maastricht
(CARIM), Maastricht University, Universiteitsingel 50, 6229
ER Maastricht, The Netherlands
| |
Collapse
|
11
|
Khan S, Rehman U, Parveen N, Kumar S, Baboota S, Ali J. siRNA therapeutics: insights, challenges, remedies and future prospects. Expert Opin Drug Deliv 2023; 20:1167-1187. [PMID: 37642354 DOI: 10.1080/17425247.2023.2251890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 08/22/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION Among conventional and novel therapeutic approaches, the siRNA strategy stands out for treating disease by silencing the gene responsible for the corresponding disorder. Gene silencing is supposedly intended to target any disease-causing gene, and therefore, several attempts and investments were made to exploit siRNA gene therapy and advance it into clinical settings. Despite the remarkable beneficial prospects, the applicability of siRNA therapeutics is very challenging due to various pathophysiological barriers that hamper its target reach, which is the cytosol, and execution of gene silencing action. AREAS COVERED The present review provides insights into the field of siRNA therapeutics, significant in vivo hurdles that mitigate the target accessibility of siRNA, and remedies to overcome these siRNA delivery challenges. Nonetheless, the current review also highlights the on-going clinical trials and the regulatory aspects of siRNA modalities. EXPERT OPINION The siRNAs have the potential to reach previously untreated target sites and silence the concerned gene owing to their modification as polymeric or lipidic nanoparticles, conjugates, and the application of advanced drug delivery strategies. With such mounting research attempts to improve the delivery of siRNA to target tissue, we might shortly witness revolutionary therapeutic outcomes, new approvals, and clinical implications.
Collapse
Affiliation(s)
- Saba Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Urushi Rehman
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Neha Parveen
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Shobhit Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut, Uttar Pradesh, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
12
|
Jiang T, Gonzalez KM, Cordova LE, Lu J. Nanotechnology-enabled gene delivery for cancer and other genetic diseases. Expert Opin Drug Deliv 2023; 20:523-540. [PMID: 37017558 PMCID: PMC10164135 DOI: 10.1080/17425247.2023.2200246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 04/04/2023] [Indexed: 04/06/2023]
Abstract
INTRODUCTION Despite gene therapy is ideal for genetic abnormality-related diseases, the easy degradation, poor targeting, and inefficiency in entering targeted cells are plaguing the effective delivery of gene therapy. Viral and non-viral vectors have been used for delivering gene therapeutics in vivo by safeguarding nucleic acid agents to target cells and to reach the specific intracellular location. A variety of nanotechnology-enabled safe and efficient systems have been successfully developed to improve the targeting ability for effective therapeutic delivery of genetic drugs. AREAS COVERED In this review, we outline the multiple biological barriers associated with gene delivery process, and highlight recent advances to gene therapy strategy in vivo, including gene correction, gene silencing, gene activation and genome editing. We point out current developments and challenges exist of non-viral and viral vector systems in association with chemical and physical gene delivery technologies and their potential for the future. EXPERT OPINION This review focuses on the opportunities and challenges to various gene therapy strategy, with specific emphasis on overcoming the challenges through the development of biocompatibility and smart gene vectors for potential clinical application.
Collapse
Affiliation(s)
- Tong Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Karina Marie Gonzalez
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Leyla Estrella Cordova
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
- NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, Arizona, 85721, United States
- BIO5 Institute, The University of Arizona, Tucson, Arizona, 85721, United States
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, 85721, United States
| |
Collapse
|
13
|
Agbaria M, Jbara-Agbaria D, Grad E, Ben-David-Naim M, Aizik G, Golomb G. Nanoparticles of VAV1 siRNA combined with LL37 peptide for the treatment of pancreatic cancer. J Control Release 2023; 355:312-326. [PMID: 36736910 DOI: 10.1016/j.jconrel.2023.01.084] [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: 11/16/2022] [Revised: 01/10/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the leading causes of cancer-related death, and it is highly resistant to therapy owing to its unique extracellular matrix. VAV1 protein, overexpressed in several cancer diseases including pancreatic cancer (PC), increases tumor proliferation and enhances metastases formation, which are associated with decreased survival. We hypothesized that an additive anti-tumor effect could be obtained by co-encapsulating in PLGA nanoparticles (NPs), the negatively charged siRNA against VAV1 (siVAV1) with the positively charged anti-tumor LL37 peptide, as a counter-ion. Several types of NPs were formulated and were characterized for their physicochemical properties, cellular internalization, and bioactivity in vitro. NPs' biodistribution, toxicity, and bioactivity were examined in a mice PDAC model. An optimal siVAV1 formulation (siVAV1-LL37 NPs) was characterized with desirable physicochemical properties in terms of nano-size, low polydispersity index (PDI), neutral surface charge, high siVAV1 encapsulation efficiency, spherical shape, and long-term shelf-life stability. Cell assays demonstrated rapid engulfment by PC cells, a specific and significant dose-dependent proliferation inhibition, as well as knockdown of VAV1 mRNA levels and migration inhibition in VAV1+ cells. Treatment with siVAV1-LL37 NPs in the mice PDAC model revealed marked accumulation of NPs in the liver and in the tumor, resulting in an increased survival rate following suppression of tumor growth and metastases, mediated via the knockdown of both VAV1 mRNA and protein levels. This proof-of-concept study validates our hypothesis of an additive effect in the treatment of PC facilitated by co-encapsulating siVAV1 in NPs with LL37 serving a dual role as a counter ion as well as an anti-tumor agent.
Collapse
Affiliation(s)
- Majd Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Doaa Jbara-Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Etty Grad
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Meital Ben-David-Naim
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Gil Aizik
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Gershon Golomb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| |
Collapse
|
14
|
Chen H, Jayasinghe MK, Yeo EYM, Wu Z, Pirisinu M, Usman WM, Pham TT, Lim KW, Tran NV, Leung AYH, Du X, Zhang Q, Phan AT, Le MTN. CD33
‐targeting extracellular vesicles deliver antisense oligonucleotides against
FLT3‐ITD
and
miR
‐125b for specific treatment of acute myeloid leukaemia. Cell Prolif 2022; 55:e13255. [PMID: 35851970 PMCID: PMC9436904 DOI: 10.1111/cpr.13255] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction Acute Myeloid Leukaemia (AML) is the most common blood cancer in adults. Although 2 out of 3 AML patients go into total remission after chemotherapies and targeted therapies, the disease recurs in 60%–65% of younger adult patients within 3 years after diagnosis with a dramatically decreased survival rate. Therapeutic oligonucleotides are promising treatments under development for AML as they can be designed to silence oncogenes with high specificity and flexibility. However, there are not many well validated approaches for safely and efficiently delivering oligonucleotide drugs. This issue could be resolved by utilizing a new generation of delivery vehicles such as extracellular vesicles (EVs). Methods In this study, we harness red blood cell‐derived EVs (RBCEVs) and engineer them via exogenous drug loading and surface functionalization to develop an efficient drug delivery system for AML. Particularly, EVs are designed to target CD33, a common surface marker with elevated expression in AML cells via the conjugation of a CD33‐binding monoclonal antibody onto the EV surface. Results The conjugation of RBCEVs with the CD33‐binding antibody significantly increases the uptake of RBCEVs by CD33‐positive AML cells, but not by CD33‐negative cells. We also load CD33‐targeting RBCEVs with antisense oligonucleotides (ASOs) targeting FLT3‐ITD or miR‐125b, 2 common oncogenes in AML, and demonstrate that the engineered EVs improve leukaemia suppression in in vitro and in vivo models of AML. Conclusion Targeted RBCEVs represent an innovative, efficient, and versatile delivery platform for therapeutic ASOs and can expedite the clinical translation of oligonucleotide drugs for AML treatments by overcoming current obstacles in oligonucleotide delivery.
Collapse
Affiliation(s)
- Huan Chen
- Department of Pharmacology, Institute for Digital Medicine, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Surgery, Immunology Program, Cancer Program and Nanomedicine Translational Program, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences City University of Hong Kong Kowloon Hong Kong SAR
| | - Migara Kavishka Jayasinghe
- Department of Pharmacology, Institute for Digital Medicine, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Surgery, Immunology Program, Cancer Program and Nanomedicine Translational Program, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Eric Yew Meng Yeo
- Department of Pharmacology, Institute for Digital Medicine, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Surgery, Immunology Program, Cancer Program and Nanomedicine Translational Program, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Zhiyuan Wu
- Department of Pharmacology, Institute for Digital Medicine, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Surgery, Immunology Program, Cancer Program and Nanomedicine Translational Program, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Marco Pirisinu
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences City University of Hong Kong Kowloon Hong Kong SAR
| | - Waqas Muhammad Usman
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences City University of Hong Kong Kowloon Hong Kong SAR
| | - Thach Tuan Pham
- Department of Pharmacology, Institute for Digital Medicine, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Surgery, Immunology Program, Cancer Program and Nanomedicine Translational Program, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Kah Wai Lim
- Division of Physics & Applied Physics, School of Physical & Mathematical Sciences Nanyang Technological University Singapore Singapore
| | - Nhan Van Tran
- Division of Physics & Applied Physics, School of Physical & Mathematical Sciences Nanyang Technological University Singapore Singapore
| | - Anskar Y. H. Leung
- Department of Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital The University of Hong Kong Pok Fu Lam Hong Kong SAR
| | - Xin Du
- Department of Hematology and Shenzhen Bone Marrow Transplantation Public Service Platform, Shenzhen Second People's Hospital The First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine Shenzhen China
| | - Qiaoxia Zhang
- Department of Hematology and Shenzhen Bone Marrow Transplantation Public Service Platform, Shenzhen Second People's Hospital The First Affiliated Hospital of Shenzhen University, Shenzhen University School of Medicine Shenzhen China
| | - Anh Tuân Phan
- Division of Physics & Applied Physics, School of Physical & Mathematical Sciences Nanyang Technological University Singapore Singapore
| | - Minh T. N. Le
- Department of Pharmacology, Institute for Digital Medicine, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Surgery, Immunology Program, Cancer Program and Nanomedicine Translational Program, Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| |
Collapse
|
15
|
A novel aptamer-based small RNA delivery platform and its application to cancer therapy. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
16
|
Jbara-Agbaria D, Blondzik S, Burger-Kentischer A, Agbaria M, Nordling-David MM, Giterman A, Aizik G, Rupp S, Golomb G. Liposomal siRNA Formulations for the Treatment of Herpes Simplex Virus-1: In Vitro Characterization of Physicochemical Properties and Activity, and In Vivo Biodistribution and Toxicity Studies. Pharmaceutics 2022; 14:633. [PMID: 35336008 PMCID: PMC8948811 DOI: 10.3390/pharmaceutics14030633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023] Open
Abstract
Herpes simplex virus-1 (HSV-1) is highly contagious, and there is a need for a therapeutic means to eradicate it. We have identified an siRNA (siHSV) that knocks down gene expression of the infected cell protein 0 (ICP0), which is important in the regulation of HSV infection. The selected siHSV was encapsulated in liposomes to overcome its poor stability, increase cell permeability, and prolonging siRNA circulation time. Several siRNAs against ICP0 have been designed and identified. We examined the role of various parameters, including formulation technique, lipids composition, and ratio. An optimal liposomal siHSV formulation (LipDOPE-siHSV) was characterized with desirable physiochemical properties, in terms of nano-size, low polydispersity index (PDI), neutral surface charge, high siHSV loading, spherical shape, high stability in physiologic conditions in vitro, and long-term shelf-life stability (>1 year, 4 °C). The liposomes exhibited profound internalization by human keratinocytes, no cytotoxicity in cell cultures, no detrimental effect on mice liver enzymes, and a gradual endo-lysosomal escape. Mice biodistribution studies in intact mice revealed accumulation, mainly in visceral organs but also in the trigeminal ganglion. The therapeutic potential of siHSV liposomes was demonstrated by significant antiviral activity both in the plaque reduction assay and in the 3D epidermis model, and the mechanism of action was validated by the reduction of ICP0 expression levels.
Collapse
Affiliation(s)
- Doaa Jbara-Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (D.J.-A.); (M.A.); (M.M.N.-D.); (A.G.); (G.A.)
| | - Saskia Blondzik
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, 70569 Stuttgart, Germany; (S.B.); (A.B.-K.); (S.R.)
| | - Anke Burger-Kentischer
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, 70569 Stuttgart, Germany; (S.B.); (A.B.-K.); (S.R.)
| | - Majd Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (D.J.-A.); (M.A.); (M.M.N.-D.); (A.G.); (G.A.)
| | - Mirjam M. Nordling-David
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (D.J.-A.); (M.A.); (M.M.N.-D.); (A.G.); (G.A.)
| | - Anna Giterman
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (D.J.-A.); (M.A.); (M.M.N.-D.); (A.G.); (G.A.)
| | - Gil Aizik
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (D.J.-A.); (M.A.); (M.M.N.-D.); (A.G.); (G.A.)
| | - Steffen Rupp
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, 70569 Stuttgart, Germany; (S.B.); (A.B.-K.); (S.R.)
| | - Gershon Golomb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (D.J.-A.); (M.A.); (M.M.N.-D.); (A.G.); (G.A.)
- The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
17
|
Kara G, Calin GA, Ozpolat B. RNAi-based therapeutics and tumor targeted delivery in cancer. Adv Drug Deliv Rev 2022; 182:114113. [PMID: 35063535 DOI: 10.1016/j.addr.2022.114113] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/15/2021] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Over the past decade, non-coding RNA-based therapeutics have proven as a great potential for the development of targeted therapies for cancer and other diseases. The discovery of the critical function of microRNAs (miRNAs) has generated great excitement in developing miRNA-based therapies. The dysregulation of miRNAs contributes to the pathogenesis of various human diseases and cancers by modulating genes that are involved in critical cellular processes, including cell proliferation, differentiation, apoptosis, angiogenesis, metastasis, drug resistance, and tumorigenesis. miRNA (miRNA mimic, anti-miRNA/antagomir) and small interfering RNA (siRNA) can inhibit the expression of any cancer-related genes/mRNAs with high specificity through RNA interference (RNAi), thus representing a remarkable therapeutic tool for targeted therapies and precision medicine. siRNA and miRNA-based therapies have entered clinical trials and recently three novel siRNA-based therapeutics were approved by the Food and Drug Administration (FDA), indicating the beginning of a new era of targeted therapeutics. The successful clinical applications of miRNA and siRNA therapeutics rely on safe and effective nanodelivery strategies for targeting tumor cells or tumor microenvironment. For this purpose, promising nanodelivery/nanoparticle-based approaches have been developed using a variety of molecules for systemic administration and improved tumor targeted delivery with reduced side effects. In this review, we present an overview of RNAi-based therapeutics, the major pharmaceutical challenges, and the perspectives for the development of promising delivery systems for clinical translation. We also highlight the passive and active tumor targeting nanodelivery strategies and primarily focus on the current applications of nanoparticle-based delivery formulations for tumor targeted RNAi molecules and their recent advances in clinical trials in human cancers.
Collapse
Affiliation(s)
- Goknur Kara
- Department of Experimental Therapeutics, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Chemistry, Biochemistry Division, Ordu University, Ordu, Turkey
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
18
|
Almarghalani DA, Boddu SHS, Ali M, Kondaka A, Ta D, Shah RA, Shah ZA. Small interfering RNAs based therapies for intracerebral hemorrhage: challenges and progress in drug delivery systems. Neural Regen Res 2022; 17:1717-1725. [PMID: 35017419 PMCID: PMC8820693 DOI: 10.4103/1673-5374.332129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke associated with higher rates of mortality. Currently, no effective drug treatment is available for ICH. The molecular pathways following ICH are complicated and diverse. Nucleic acid therapeutics such as gene knockdown by small interfering RNAs (siRNAs) have been developed in recent years to modulate ICH’s destructive pathways and mitigate its outcomes. However, siRNAs delivery to the central nervous system is challenging and faces many roadblocks. Existing barriers to systemic delivery of siRNA limit the use of naked siRNA; therefore, siRNA-vectors developed to protect and deliver these therapies into the specific-target areas of the brain, or cell types seem quite promising. Efficient delivery of siRNA via nanoparticles emerged as a viable and effective alternative therapeutic tool for central nervous system-related diseases. This review discusses the obstacles to siRNA delivery, including the advantages and disadvantages of viral and nonviral vectors. Additionally, we provide a comprehensive overview of recent progress in nanotherapeutics areas, primarily focusing on the delivery system of siRNA for ICH treatment.
Collapse
Affiliation(s)
- Daniyah A Almarghalani
- Department of Pharmacology and Experimental Therapeutics; Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Mohammad Ali
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Akhila Kondaka
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Devin Ta
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Rayyan A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
19
|
Biber G, Sabag B, Raiff A, Ben‐Shmuel A, Puthenveetil A, Benichou JIC, Jubany T, Levy M, Killner S, Barda‐Saad M. Modulation of intrinsic inhibitory checkpoints using nano-carriers to unleash NK cell activity. EMBO Mol Med 2022; 14:e14073. [PMID: 34725941 PMCID: PMC8749471 DOI: 10.15252/emmm.202114073] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/22/2021] [Accepted: 09/30/2021] [Indexed: 01/22/2023] Open
Abstract
Natural killer (NK) cells provide a powerful weapon mediating immune defense against viral infections, tumor growth, and metastatic spread. NK cells demonstrate great potential for cancer immunotherapy; they can rapidly and directly kill cancer cells in the absence of MHC-dependent antigen presentation and can initiate a robust immune response in the tumor microenvironment (TME). Nevertheless, current NK cell-based immunotherapies have several drawbacks, such as the requirement for ex vivo expansion of modified NK cells, and low transduction efficiency. Furthermore, to date, no clinical trial has demonstrated a significant benefit for NK-based therapies in patients with advanced solid tumors, mainly due to the suppressive TME. To overcome current obstacles in NK cell-based immunotherapies, we describe here a non-viral lipid nanoparticle-based delivery system that encapsulates small interfering RNAs (siRNAs) to gene silence the key intrinsic inhibitory NK cell molecules, SHP-1, Cbl-b, and c-Cbl. The nanoparticles (NPs) target NK cells in vivo, silence inhibitory checkpoint signaling molecules, and unleash NK cell activity to eliminate tumors. Thus, the novel NP-based system developed here may serve as a powerful tool for future NK cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Guy Biber
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Batel Sabag
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Anat Raiff
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Aviad Ben‐Shmuel
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Abhishek Puthenveetil
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Jennifer I C Benichou
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Tammir Jubany
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Moria Levy
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Shiran Killner
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Mira Barda‐Saad
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| |
Collapse
|
20
|
Cerqueni G, Scalzone A, Licini C, Gentile P, Mattioli-Belmonte M. Insights into oxidative stress in bone tissue and novel challenges for biomaterials. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 130:112433. [PMID: 34702518 DOI: 10.1016/j.msec.2021.112433] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/28/2022]
Abstract
The presence of Reactive Oxygen Species (ROS) in bone can influence resident cells behaviour as well as the extra-cellular matrix composition and the tissue architecture. Aging, in addition to excessive overloads, unbalanced diet, smoking, predisposing genetic factors, lead to an increase of ROS and, if it is accompanied with an inappropriate production of scavengers, promotes the generation of oxidative stress that encourages bone catabolism. Furthermore, bone injuries can be triggered by numerous events such as road and sports accidents or tumour resection. Although bone tissue possesses a well-known repair and regeneration capacity, these mechanisms are inefficient in repairing large size defects and bone grafts are often necessary. ROS play a fundamental role in response after the implant introduction and can influence its success. This review provides insights on the mechanisms of oxidative stress generated by an implant in vivo and suitable ways for its modulation. The local delivery of active molecules, such as polyphenols, enhanced bone biomaterial integration evidencing that the management of the oxidative stress is a target for the effectiveness of an implant. Polyphenols have been widely used in medicine for cardiovascular, neurodegenerative, bone disorders and cancer, thanks to their antioxidant and anti-inflammatory properties. In addition, the perspective of new smart biomaterials and molecular medicine for the oxidative stress modulation in a programmable way, by the use of ROS responsive materials or by the targeting of selective molecular pathways involved in ROS generation, will be analysed and discussed critically.
Collapse
Affiliation(s)
- Giorgia Cerqueni
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy
| | - Annachiara Scalzone
- School of Engineering, Newcastle University, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Caterina Licini
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy; Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 204, 10129 Torino, Italy
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60126, Italy.
| |
Collapse
|
21
|
Bogusławska-Duch J, Ducher-Hanaka M, Zajkowska A, Czajka M, Małecki M. Therapeutic combination silencing VEGF and SOX10 increases the antiangiogenic effect in the mouse melanoma model B16-F10 - in vitro and in vivo studies. Postepy Dermatol Alergol 2021; 38:887-898. [PMID: 34849139 PMCID: PMC8610042 DOI: 10.5114/ada.2021.110461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/24/2020] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Gene therapy is an innovative form of treatment of genetic diseases, in which psiRNA molecules silencing specific genes are applied. AIM The study evaluated the anti-tumour effect of psiRNA silencing preparations of the vascular endothelial growth factor (VEGF) and Sry-related HMG-Box gene 10 (SOX10) on melanoma (B16-F10) by inhibiting angiogenesis. MATERIAL AND METHODS The preparations based on plasmid vectors psiRNA silencing the gene SOX10 and VEGF that form complexes with cationic lipid (psiRNA/carrier) have been developed. psiRNA preparations were tested on the mouse melanoma cell line B16-F10, both in vitro and in vivo. The silencing activity of transfected melanoma cells with the obtained psiRNA preparations was examined using the qPCR and Western blot methods. The anti-tumour activity of psiRNA preparations on melanoma tumour cells was then evaluated in a mouse in vivo model. RESULTS In vitro studies have shown that the B16-F10 cells efficiently transfect non-viral preparations - psiRNA: Lyovec (74-89%). Worth mentioning is the fact that silencing SOX10 in B16-F10 melanoma cells increases the expression of the COL18A1 gene (compared to the preparation inhibiting only VEGF), which codes the endostatin to stop angiogenesis. In vivo results show that the level of haemoglobin in tumours of mice treated with psiRNA formulations was over 6 times lower than controls and tumour mass was 60-80% lower. CONCLUSIONS The novel study proves that simultaneous inhibition of SOX10 and VEGF enhances the antiangiogenic action and thus contributes to a significant halt of disease development. In addition, these data expand knowledge about SOX10 regulation and functions.
Collapse
Affiliation(s)
| | | | - Agnieszka Zajkowska
- Department of Applied Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Milena Czajka
- Department of Applied Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Montinari MR, Minelli S, De Caterina R. Eighty years of oral anticoagulation: Learning from history. Vascul Pharmacol 2021; 141:106918. [PMID: 34537376 DOI: 10.1016/j.vph.2021.106918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 02/05/2023]
Abstract
In the year 2021 we celebrate the 80th anniversary of the first clinical use of vitamin K antagonists (VKAs), the mainstay of prevention and long-term treatment of thromboembolic disease. The discovery and development of oral anticoagulants is one of the most important chapters in the history of medicine, a goal pursued by physicians trying to combat the clinical manifestations of thrombosis since ancient times. Until the last decade, VKAs were the only oral anticoagulants available and used in clinical practice. Today, their clinical use has progressively shrunk, as the non-vitamin K antagonist oral anticoagulants (NOACs) are increasingly replacing VKAs in various conditions after the successful completion of several large randomized controlled trials. Currently, new research is tackling upstream components of the intrinsic pathway - particularly factor XI and factor XII - for the development of new, even safer anticoagulants promising to reduce bleeding without compromising efficacy. This review highlights the evolution of oral anticoagulant therapy tracing the key stages of a long and fascinating history that has unfolded from the first part of the twentieth century until today, indeed an intriguing journey where serendipity is intertwined with the tenacious work of many researchers.
Collapse
Affiliation(s)
- Maria Rosa Montinari
- Chair of History of Medicine, Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
| | | | - Raffaele De Caterina
- Chair of Cardiology, University of Pisa, University Cardiology Division, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Fondazione VillaSerena, Città Sant'Angelo, Pescara, Italy.
| |
Collapse
|
23
|
Kampel L, Goldsmith M, Ramishetti S, Veiga N, Rosenblum D, Gutkin A, Chatterjee S, Penn M, Lerman G, Peer D, Muhanna N. Therapeutic inhibitory RNA in head and neck cancer via functional targeted lipid nanoparticles. J Control Release 2021; 337:378-389. [PMID: 34303750 DOI: 10.1016/j.jconrel.2021.07.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 07/07/2021] [Accepted: 07/19/2021] [Indexed: 12/25/2022]
Abstract
Currently there are no specific therapies addressing the distinctive biology of human papillomavirus (HPV)-induced cancer approved for clinical use. Short interfering RNA (siRNA) has much potential for therapeutic manipulation of HPV E6/E7 oncoproteins. Lipid-based nanoparticles (LNPs) can be utilized for systemic transportation and delivery of siRNA at target site. We recently developed a recombinant protein linker that enables uniform conjugation of targeting antibodies to the LNPs. Herein, we demonstrate the therapeutic efficacy of anti-E6/E7 siRNA delivered via targeted LNPs (tLNPs) in a xenograft HPV-positive tumor model. We show that anti-epidermal growth factor receptor (EGFR) antibodies, anchored to the LNPs as targeting moieties, facilitate cargo delivery but also mediate anti-tumor activity. Treatment with siE6 via tLNPs resulted in 50% greater reduction of tumor volume compared to treatment with siControl encapsulated in isoLNPs (coated with isotype control antibodies). We demonstrate superior suppression of HPV oncogenes and higher induction of apoptosis by the tLNPs both in vitro and in vivo. Altogether, the coupling of inhibitory siE6 with anti-EGFR antibodies, that further elicited anti-tumor effects, successfully restricted tumor progression. This system that combines potent siRNA and therapeutically functional tLNPs can be modulated against various cancer models.
Collapse
Affiliation(s)
- Liyona Kampel
- The Head and Neck Cancer Research Laboratory, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel; The Department of Otolaryngology, Head and Neck Surgery and Maxillofacial Surgery, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel; Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Meir Goldsmith
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Srinivas Ramishetti
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nuphar Veiga
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniel Rosenblum
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Gutkin
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Sushmita Chatterjee
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel
| | - Moran Penn
- The Head and Neck Cancer Research Laboratory, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel
| | - Galya Lerman
- The Head and Neck Cancer Research Laboratory, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Tel Aviv University, Tel Aviv 69978, Israel; Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Department of Materials Sciences & Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 69978, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Nidal Muhanna
- The Head and Neck Cancer Research Laboratory, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel; The Department of Otolaryngology, Head and Neck Surgery and Maxillofacial Surgery, Tel-Aviv Sourasky Medical Center, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6423906, Israel.
| |
Collapse
|
24
|
Liu Y, Zhu X, Wei Z, Feng W, Li L, Ma L, Li F, Zhou J. Customized Photothermal Therapy of Subcutaneous Orthotopic Cancer by Multichannel Luminescent Nanocomposites. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2008615. [PMID: 34121241 DOI: 10.1002/adma.202008615] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/07/2021] [Indexed: 06/12/2023]
Abstract
Photothermal therapy (PTT) is a potentially advanced strategy for highly precise cancer treatment. Tumor-microenvironment-activatable agents provide useful tools for PTT, but their photothermal conversion capacities vary and cannot be evaluated in vivo; thus, a general PTT prescription does not work with individual activatable agents. Here, glutathione (GSH)-activatable nanocomposites, silicomolybdate-functionalized NaLuF4 :Yb,Er@NaLuF4 @NaLuF4 :Nd are prepared, for customized PTT of subcutaneous orthotopic cancer. By simultaneously determining intratumoral GSH concentration and the amount of accumulated agent using multiple orthogonal luminescent emissions of nanocomposites, near-infrared absorbance of photothermal conversion agents is evaluated in vivo, based on the optimized irradiating prescriptions (irradiating power density and time) established. This allows customized PTT of each individual case with high efficacy and viability. This work also includes a method for investigating individual intratumoral variation, and the development of the next generation of customized nanomedicine for efficacious PTT of subcutaneous orthotopic cancer.
Collapse
Affiliation(s)
- Yuxin Liu
- Beijing Key Laboratory for Optical Materials and Photonic Devices & Department of Chemistry, Capital Normal University, Beijing, 100048, China
- Department of Biomolecular System, Max-Planck Institute for Colloids and Interfaces, 14476, Potsdam, Germany
| | - Xingjun Zhu
- Department of Chemistry & Institutes of Biomedical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
- School of Physical Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Zheng Wei
- Beijing Key Laboratory for Optical Materials and Photonic Devices & Department of Chemistry, Capital Normal University, Beijing, 100048, China
| | - Wei Feng
- Department of Chemistry & Institutes of Biomedical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Luoyuan Li
- Beijing Key Laboratory for Optical Materials and Photonic Devices & Department of Chemistry, Capital Normal University, Beijing, 100048, China
| | - Liyi Ma
- Beijing Key Laboratory for Optical Materials and Photonic Devices & Department of Chemistry, Capital Normal University, Beijing, 100048, China
| | - Fuyou Li
- Department of Chemistry & Institutes of Biomedical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Jing Zhou
- Beijing Key Laboratory for Optical Materials and Photonic Devices & Department of Chemistry, Capital Normal University, Beijing, 100048, China
| |
Collapse
|
25
|
Carvalho BG, Vit FF, Carvalho HF, Han SW, de la Torre LG. Recent advances in co-delivery nanosystems for synergistic action in cancer treatment. J Mater Chem B 2021; 9:1208-1237. [PMID: 33393582 DOI: 10.1039/d0tb02168g] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nanocarrier delivery systems have been widely studied to carry unique or dual chemical drugs. The major challenge of chemotherapies is to overcome the multidrug-resistance (MDR) of cells to antineoplastic medicines. In this context, nano-scale technology has allowed researchers to develop biocompatible nano-delivery systems to overcome the limitation of chemical agents. The development of nano-vehicles may also be directed to co-deliver different agents such as drugs and genetic materials. The delivery of nucleic acids targeting specific cells is based on gene therapy principles to replace the defective gene, correct genome errors or knock-down a particular gene. Co-delivery systems are attractive strategies due to the possibility of achieving synergistic therapeutic effects, which are more effective in overcoming the MDR of cancer cells. These combined therapies can provide better outcomes than separate delivery approaches carrying either siRNA, miRNA, pDNA, or drugs. This article reviews the main design features that need to be associated with nano-vehicles to co-deliver drugs, genes, and gene-drug combinations with efficacy. The advantages and disadvantages of co-administration approaches are also overviewed and compared with individual nanocarrier systems. Herein, future trends and perspectives in designing novel nano-scale platforms to co-deliver therapeutic agents are also discussed.
Collapse
Affiliation(s)
- Bruna G Carvalho
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas, Brazil.
| | - Franciele F Vit
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas, Brazil.
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Sang W Han
- Department of Biophysics, Federal University of São Paulo, Center for Cell and Molecular Therapy, São Paulo, Brazil
| | - Lucimara G de la Torre
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas, Brazil.
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW The development of mRNA vaccines against coronavirus disease 2019 has brought worldwide attention to the transformative potential of RNA-based therapeutics. The latter is essentially biological software that can be rapidly designed and generated, with an extensive catalog of applications. This review aims to highlight the mechanisms of action by which RNA-based drugs can affect specific gene targets and how RNA drugs can be employed to treat cardiovascular disease, with the focus on the therapeutics being evaluated in clinical trials. The recent advances in nanotechnology aiding the translation of such therapies into the clinic are also discussed. RECENT FINDINGS There is a growing body of studies demonstrating utility of RNA for targeting previously 'undruggable' pathways involved in development and progression of cardiovascular disease. Some challenges in RNA delivery have been overcome thanks to nanotechnology. There are several RNA-based drugs to treat hypercholesterolemia and myocardial infarction which are currently in clinical trials. SUMMARY RNA therapeutics is a rapidly emerging field of biotherapeutics based upon a powerful and versatile platform with a nearly unlimited capacity to address unmet clinical needs. These therapeutics are destined to change the standard of care for many diseases, including cardiovascular disease.
Collapse
Affiliation(s)
- Christian Boada
- RNA Therapeutics Program, Department of Cardiovascular Sciences, Houston Methodist Academic Institute, TX
- Texas A&M School of Medicine, College Station, TX
| | - Roman Sukhovershin
- RNA Therapeutics Program, Department of Cardiovascular Sciences, Houston Methodist Academic Institute, TX
| | | | - John P. Cooke
- RNA Therapeutics Program, Department of Cardiovascular Sciences, Houston Methodist Academic Institute, TX
| |
Collapse
|
27
|
Zhu YX, Jia HR, Duan QY, Wu FG. Nanomedicines for combating multidrug resistance of cancer. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1715. [PMID: 33860622 DOI: 10.1002/wnan.1715] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Chemotherapy typically involves the use of specific chemodrugs to inhibit the proliferation of cancer cells, but the frequent emergence of a variety of multidrug-resistant cancer cells poses a tremendous threat to our combat against cancer. The fundamental causes of multidrug resistance (MDR) have been studied for decades, and can be generally classified into two types: one is associated with the activation of diverse drug efflux pumps, which are responsible for translocating intracellular drug molecules out of the cells; the other is linked with some non-efflux pump-related mechanisms, such as antiapoptotic defense, enhanced DNA repair ability, and powerful antioxidant systems. To overcome MDR, intense efforts have been made to develop synergistic therapeutic strategies by introducing MDR inhibitors or combining chemotherapy with other therapeutic modalities, such as phototherapy, gene therapy, and gas therapy, in the hope that the drug-resistant cells can be sensitized toward chemotherapeutics. In particular, nanotechnology-based drug delivery platforms have shown the potential to integrate multiple therapeutic agents into one system. In this review, the focus was on the recent development of nanostrategies aiming to enhance the efficiency of chemotherapy and overcome the MDR of cancer in a synergistic manner. Different combinatorial strategies are introduced in detail and the advantages as well as underlying mechanisms of why these strategies can counteract MDR are discussed. This review is expected to shed new light on the design of advanced nanomedicines from the angle of materials and to deepen our understanding of MDR for the development of more effective anticancer strategies. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Ya-Xuan Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Hao-Ran Jia
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Qiu-Yi Duan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
28
|
Spencer DS, Shodeinde AB, Beckman DW, Luu BC, Hodges HR, Peppas NA. Cytocompatibility, membrane disruption, and siRNA delivery using environmentally responsive cationic nanogels. J Control Release 2021; 332:608-619. [PMID: 33675879 PMCID: PMC8089052 DOI: 10.1016/j.jconrel.2021.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 11/17/2022]
Abstract
Advances in the formulation of nucleic acid-based therapeutics have rendered them a promising avenue for treating diverse ailments. Nonetheless, clinical translation of these therapies is hindered by a lack of strategies to ensure the delivery of these nucleic acids in a safe, efficacious manner with the required spatial and temporal control. To this aim, environmentally responsive hydrogels are of interest due to their ability to provide the desired characteristics of a protective carrier for siRNA delivery. Previous work in our laboratory has demonstrated the ability to synthesize nanoparticle formulations with targeted pKa, swelling, and surface PEG density. Here, a library of nanoparticle formulations was assessed on their in vitro toxicity, hemolytic capacity, siRNA loading, and gene-silencing efficacy. Successful candidates exhibited the lowest degrees of cytotoxicity, pH-dependent membrane disruption potential, the highest siRNA loading, and the highest transfection efficacies.
Collapse
Affiliation(s)
- David S Spencer
- 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
| | - David W Beckman
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX 78712, USA
| | - Bryan C Luu
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX 78712, USA
| | - Hannah R Hodges
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX 78712, USA
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, 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; 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; Departments of Pediatrics, Surgery, and Perioperative Care, Dell Medical School, 1601 Trinity St., Bldg. B, Stop Z0800, Austin, TX 78712, USA.
| |
Collapse
|
29
|
A novel rationale for targeting FXI: Insights from the hemostatic microRNA targetome for emerging anticoagulant strategies. Pharmacol Ther 2021; 218:107676. [DOI: 10.1016/j.pharmthera.2020.107676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
|
30
|
Lin G, Revia RA, Zhang M. Inorganic Nanomaterial-Mediated Gene Therapy in Combination with Other Antitumor Treatment Modalities. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2007096. [PMID: 34366761 PMCID: PMC8336227 DOI: 10.1002/adfm.202007096] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Indexed: 05/05/2023]
Abstract
Cancer is a genetic disease originating from the accumulation of gene mutations in a cellular subpopulation. Although many therapeutic approaches have been developed to treat cancer, recent studies have revealed an irrefutable challenge that tumors evolve defenses against some therapies. Gene therapy may prove to be the ultimate panacea for cancer by correcting the fundamental genetic errors in tumors. The engineering of nanoscale inorganic carriers of cancer therapeutics has shown promising results in the efficacious and safe delivery of nucleic acids to treat oncological diseases in small-animal models. When these nanocarriers are used for co-delivery of gene therapeutics along with auxiliary treatments, the synergistic combination of therapies often leads to an amplified health benefit. In this review, an overview of the inorganic nanomaterials developed for combinatorial therapies of gene and other treatment modalities is presented. First, the main principles of using nucleic acids as therapeutics, inorganic nanocarriers for medical applications and delivery of gene/drug payloads are introduced. Next, the utility of recently developed inorganic nanomaterials in different combinations of gene therapy with each of chemo, immune, hyperthermal, and radio therapy is examined. Finally, current challenges in the clinical translation of inorganic nanomaterial-mediated therapies are presented and outlooks for the field are provided.
Collapse
Affiliation(s)
- Guanyou Lin
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Richard A Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
31
|
Shodeinde AB, Murphy AC, Oldenkamp HF, Potdar AS, Ludolph CM, Peppas NA. Recent Advances in Smart Biomaterials for the Detection and Treatment of Autoimmune Diseases. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909556. [PMID: 33071713 PMCID: PMC7566744 DOI: 10.1002/adfm.201909556] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/15/2020] [Indexed: 05/07/2023]
Abstract
Autoimmune diseases are a group of debilitating illnesses that are often idiopathic in nature. The steady rise in the prevalence of these conditions warrants new approaches for diagnosis and treatment. Stimuli-responsive biomaterials also known as "smart", "intelligent" or "recognitive" biomaterials are widely studied for their applications in drug delivery, biosensing and tissue engineering due to their ability to produce thermal, optical, chemical, or structural changes upon interacting with the biological environment. This critical analysis highlights studies within the last decade that harness the recognitive capabilities of these biomaterials towards the development of novel detection and treatment options for autoimmune diseases.
Collapse
Affiliation(s)
- Aaliyah B. Shodeinde
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Andrew C. Murphy
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Heidi F. Oldenkamp
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Abhishek S. Potdar
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
| | - Catherine M. Ludolph
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
| | - Nicholas A. Peppas
- McKetta Department of Chemical Engineering, 200 E. Dean Keeton St. Stop C0400, Austin, TX, USA, 78712
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX, USA, 78712
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave. Stop A1900, Austin, TX, USA, 78712
- Department of Surgery and Perioperative Care, Dell Medical School, 1601 Trinity St., Bldg. B, Stop Z0800, Austin, TX, USA, 78712
- Department of Pediatrics, Dell Medical School, 1400 Barbara Jordan Blvd., Austin, TX, USA, 78723
| |
Collapse
|
32
|
Cunha-Santos C, Perdigao PRL, Martin F, Oliveira JG, Cardoso M, Manuel A, Taveira N, Goncalves J. Inhibition of HIV replication through siRNA carried by CXCR4-targeted chimeric nanobody. Cell Mol Life Sci 2020; 77:2859-2870. [PMID: 31641784 PMCID: PMC11104913 DOI: 10.1007/s00018-019-03334-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/24/2019] [Accepted: 10/03/2019] [Indexed: 01/05/2023]
Abstract
Small interfering RNA (siRNA) application in therapy still faces a major challenge with the lack of an efficient and specific delivery system. Current vehicles are often responsible for poor efficacy, safety concerns, and burden costs of siRNA-based therapeutics. Here, we describe a novel strategy for targeted delivery of siRNA molecules to inhibit human immunodeficiency virus (HIV) infection. Specific membrane translocation of siRNA inhibitor was addressed by an engineered nanobody targeting the HIV co-receptor CXCR4 (NbCXCR4) in fusion with a single-chain variable fragment (4M5.3) that carried the FITC-conjugated siRNA. 4M5.3-NbCXCR4 conjugate (4M5.3X4) efficiently targeted CXCR4+ T lymphocytes, specifically translocating siRNA by receptor-mediated endocytosis. Targeted delivery of siRNA directed to the mRNA of HIV transactivator tat silenced Tat-driven viral transcription and inhibited the replication of distinct virus clades. In summary, we have shown that the engineered nanobody chimera developed in this study constitutes an efficient and specific delivery method of siRNAs through CXCR4 receptor.
Collapse
Affiliation(s)
- Catarina Cunha-Santos
- Molecular Microbiology and Biotechnology Department, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Pedro Ricardo Lucas Perdigao
- Molecular Microbiology and Biotechnology Department, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Francisco Martin
- HIV Evolution, Epidemiology and Prevention Department, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana Gomes Oliveira
- Molecular Microbiology and Biotechnology Department, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Miguel Cardoso
- Molecular Microbiology and Biotechnology Department, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Ana Manuel
- Molecular Microbiology and Biotechnology Department, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Nuno Taveira
- HIV Evolution, Epidemiology and Prevention Department, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, Monte de Caparica, Portugal
| | - Joao Goncalves
- Molecular Microbiology and Biotechnology Department, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
33
|
Kim T, Viard M, Afonin KA, Gupta K, Popov M, Salotti J, Johnson PF, Linder C, Heldman E, Shapiro BA. Characterization of Cationic Bolaamphiphile Vesicles for siRNA Delivery into Tumors and Brain. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:359-372. [PMID: 32200271 PMCID: PMC7090283 DOI: 10.1016/j.omtn.2020.02.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/19/2019] [Accepted: 02/23/2020] [Indexed: 12/27/2022]
Abstract
Small interfering RNAs (siRNAs) are potential therapeutic substances due to their gene silencing capability as exemplified by the recent approval by the US Food and Drug Administration (FDA) of the first siRNA therapeutic agent (patisiran). However, the delivery of naked siRNAs is challenging because of their short plasma half-lives and poor cell penetrability. In this study, we used vesicles made from bolaamphiphiles (bolas), GLH-19 and GLH-20, to investigate their ability to protect siRNA from degradation by nucleases while delivering it to target cells, including cells in the brain. Based on computational and experimental studies, we found that GLH-19 vesicles have better delivery characteristics than do GLH-20 vesicles in terms of stability, binding affinity, protection against nucleases, and transfection efficiency, while GLH-20 vesicles contribute to efficient release of the delivered siRNAs, which become available for silencing. Our studies with vesicles made from a mixture of the two bolas (GLH-19 and GLH-20) show that they were able to deliver siRNAs into cultured cancer cells, into a flank tumor and into the brain. The vesicles penetrate cell membranes and the blood-brain barrier (BBB) by endocytosis and transcytosis, respectively, mainly through the caveolae-dependent pathway. These results suggest that GLH-19 strengthens vesicle stability, provides protection against nucleases, and enhances transfection efficiency, while GLH-20 makes the siRNA available for gene silencing.
Collapse
Affiliation(s)
- Taejin Kim
- RNA Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Mathias Viard
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kirill A Afonin
- Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; The Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Kshitij Gupta
- RNA Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Mary Popov
- Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jacqueline Salotti
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Peter F Johnson
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | - Bruce A Shapiro
- RNA Biology Laboratory, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
34
|
Veiga N, Diesendruck Y, Peer D. Targeted lipid nanoparticles for RNA therapeutics and immunomodulation in leukocytes. Adv Drug Deliv Rev 2020; 159:364-376. [PMID: 32298783 DOI: 10.1016/j.addr.2020.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/27/2020] [Accepted: 04/10/2020] [Indexed: 12/25/2022]
Abstract
Abnormalities in leukocytes' function are associated with many immune related disorders, such as cancer, autoimmunity and susceptibility to infectious diseases. Recent developments in Genome-wide-association-studies give rise to new opportunities for novel therapeutics. RNA-based modalities, that allow a selective genetic manipulation in vivo, are powerful tools for personalized medicine, enabling downregulation or expression of relevant proteins. Yet, RNA-based therapeutics requires a delivery modality to facilitate the stability, uptake and intracellular release of the RNA molecules. The use of lipid nanoparticles as a drug delivery approach improves the payloads' stability, pharmacokinetics, bio-distribution and therapeutic benefit while reducing side effects. Moreover, a wide variety of targeting moieties allow a precise and modular manipulation of gene expression, together with the ability to identify and selectively affect disease-relevant leukocytes-subsets. Altogether, RNA-based therapeutics, targeting leukocytes subsets, is believed to be one of the most promising therapeutic concepts of the near future, addressing pressing issues in cancer and inflammation heterogeneity.
Collapse
|
35
|
Adir O, Poley M, Chen G, Froim S, Krinsky N, Shklover J, Shainsky-Roitman J, Lammers T, Schroeder A. Integrating Artificial Intelligence and Nanotechnology for Precision Cancer Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901989. [PMID: 31286573 PMCID: PMC7124889 DOI: 10.1002/adma.201901989] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/17/2019] [Indexed: 05/13/2023]
Abstract
Artificial intelligence (AI) and nanotechnology are two fields that are instrumental in realizing the goal of precision medicine-tailoring the best treatment for each cancer patient. Recent conversion between these two fields is enabling better patient data acquisition and improved design of nanomaterials for precision cancer medicine. Diagnostic nanomaterials are used to assemble a patient-specific disease profile, which is then leveraged, through a set of therapeutic nanotechnologies, to improve the treatment outcome. However, high intratumor and interpatient heterogeneities make the rational design of diagnostic and therapeutic platforms, and analysis of their output, extremely difficult. Integration of AI approaches can bridge this gap, using pattern analysis and classification algorithms for improved diagnostic and therapeutic accuracy. Nanomedicine design also benefits from the application of AI, by optimizing material properties according to predicted interactions with the target drug, biological fluids, immune system, vasculature, and cell membranes, all affecting therapeutic efficacy. Here, fundamental concepts in AI are described and the contributions and promise of nanotechnology coupled with AI to the future of precision cancer medicine are reviewed.
Collapse
Affiliation(s)
- Omer Adir
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
- The Norman Seiden Multidisciplinary Program for Nanoscience and Nanotechnology, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Maria Poley
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Gal Chen
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Sahar Froim
- Department of Physical Electronics, School of Electrical Engineering, Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Nitzan Krinsky
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Jeny Shklover
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Janna Shainsky-Roitman
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, 52074, Germany
| | - Avi Schroeder
- Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| |
Collapse
|
36
|
Molla MR, Chakraborty S, Munoz Sagredo L, Drechsler M, Orian Rousseau V, Levkin PA. Combinatorial Synthesis of a Lipidoid Library by Thiolactone Chemistry: In Vitro Screening and In Vivo Validation for siRNA Delivery. Bioconjug Chem 2020; 31:852-860. [PMID: 32068393 DOI: 10.1021/acs.bioconjchem.0c00013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transcriptional inhibition by small interfering RNA (siRNA) delivery using synthetic transfection agents eliminates the subsequent risk of introducing mutations in relevant genes, as opposed to viral vectors. However, synthetic vectors with comparable transfection efficiency to that of viral vectors are yet to be developed. Hence, synthesizing new transfection vehicles with low toxicity is important. In this study, a library of lipid-like molecules (lipidoids) was synthesized by thiolactone chemistry. This library facilitated nonviral delivery of siRNA to mammalian cells, inducing sequence-specific knockdown of a target gene. The liposomal nanoparticles complexed with anti-green fluorescent protein (GFP) siRNA were successfully screened for transfection efficiency using a HeLa-GFP cell line. The five best-performing lipidoids identified in the screening were found to exhibit superior GFP-knockdown efficiency compared with commercially available transfection reagents. The efficiency of siRNA delivery by one of these lipidoids with minimal toxicity was further successfully evaluated in vivo using Kdrl:EGFP zebrafish embryos as a model system. Our study would be important as a facile synthetic route of efficient nonviral nucleic acid delivery to live cells and organisms.
Collapse
Affiliation(s)
- Mijanur R Molla
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany.,Department of Chemistry, University of Calcutta (Rashbehari Siksha Prangan), 92 A. P. C. Road, Kolkata 700009, India
| | - Shraddha Chakraborty
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Leonel Munoz Sagredo
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany.,Faculty of Medicine, University of Valparaiso, Hontaneda 2653, 2341369 Valparaiso, Chile
| | - Markus Drechsler
- Bavarian Polymer Institute (BPI), University of Bayreuth, Universitaetsstr. 30, D-95440 Bayreuth, Germany
| | - Véronique Orian Rousseau
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Pavel A Levkin
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
37
|
Borgheti-Cardoso LN, Viegas JSR, Silvestrini AVP, Caron AL, Praça FG, Kravicz M, Bentley MVLB. Nanotechnology approaches in the current therapy of skin cancer. Adv Drug Deliv Rev 2020; 153:109-136. [PMID: 32113956 DOI: 10.1016/j.addr.2020.02.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/16/2019] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
Skin cancer is a high burden disease with a high impact on global health. Conventional therapies have several drawbacks; thus, the development of effective therapies is required. In this context, nanotechnology approaches are an attractive strategy for cancer therapy because they enable the efficient delivery of drugs and other bioactive molecules to target tissues with low toxic effects. In this review, nanotechnological tools for skin cancer will be summarized and discussed. First, pathology and conventional therapies will be presented, followed by the challenges of skin cancer therapy. Then, the main features of developing efficient nanosystems will be discussed, and next, the most commonly used nanoparticles (NPs) described in the literature for skin cancer therapy will be presented. Subsequently, the use of NPs to deliver chemotherapeutics, immune and vaccine molecules and nucleic acids will be reviewed and discussed as will the combination of physical methods and NPs. Finally, multifunctional delivery systems to codeliver anticancer therapeutic agents containing or not surface functionalization will be summarized.
Collapse
|
38
|
Chandra S, Michael Nguyen H, Wiltz K, Hall N, Chaudhry S, Olverson G, Mandal T, Dash S, Kundu A. Aptamer-functionalized Hybrid Nanoparticles to Enhance the Delivery of Doxorubicin into Breast Cancer Cells by Silencing P-glycoprotein. JOURNAL OF CANCER TREATMENT & DIAGNOSIS 2020; 4:1-13. [PMID: 32395707 PMCID: PMC7213597 DOI: 10.29245/2578-2967/2020/1.1176] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The MDR of metastatic breast cancer cells is accompanied by the overexpression of P-gp transporter. This study has been focused to determine whether silencing the expression of P-gp by aptamer-labeled siRNA nanoparticles could enhance the delivery of doxorubicin into breast cancer cells in culture. METHODOLOGY The nanoparticle F-31 was prepared using DOTAP, cholesterol, and PLGA, and then incorporating Mal-PEG to facilitate aptamer-binding. The nanoparticles were surface-functionalized with aptamer A6, which targets Her-2 receptors overexpressed on the surface of breast cancer cells. RESULTS This study has shown that the uptake of Dox by Dox-resistant 4T1-R is significantly less than Dox-sensitive 4T1-S which is partly attributed to the higher expression of drug-efflux pump P-gp on the surface of the resistant cells. The targeted knockdown of P-gp has been enhanced when the particles carrying P-gp siRNA was labeled with aptamer. Concurrently, the uptake of Dox into the Dox-resistant 4T1-R breast cancer cells has increased significantly when the P-gp was silenced by P-gp siRNA-encapsulated aptamer-labeled nanoparticles. CONCLUSIONS This preliminary study concludes that downregulating P-gp expression by targeted delivery of P-gp siRNA using aptamer-labeled lipid-based hybrid nanoparticles could effectively increase the intracellular trafficking of doxorubicin in Dox-resistant mouse breast cancer cells.
Collapse
Affiliation(s)
- Sruti Chandra
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana
| | | | - Kylar Wiltz
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana
| | - Nicholas Hall
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana
| | - Shanzay Chaudhry
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana
| | - George Olverson
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana
| | - Tarun Mandal
- Center for Nanomedicine and Drug Delivery, Xavier University College of Pharmacy, New Orleans, Louisiana
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Anup Kundu
- Department of Biology, Xavier University of Louisiana, New Orleans, Louisiana
| |
Collapse
|
39
|
Formulation of RNA interference-based drugs for pulmonary delivery: challenges and opportunities. Ther Deliv 2019; 9:731-749. [PMID: 30277138 DOI: 10.4155/tde-2018-0029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
With recent advances in the field of RNAi-based therapeutics, it is possible to make any target gene 'druggable', at least in principle. The present review focuses on aspects critical for pulmonary delivery of formulations of nucleic acid-based drugs. The first part introduces the therapeutic potential of RNAi-based drugs for the treatment of lung diseases. Subsequently, we discuss opportunities for formulation-enabled pulmonary delivery of RNAi drugs in light of key physicochemical properties and physiological barriers. In the following section, an overview is included of methodologies for imparting inhalable characteristics to nucleic acid formulations. Finally, we review one of the bottlenecks in the early preclinical testing of inhalable nucleic acid-based formulations, in other words, devices suitable for pulmonary administration of powder-based formulations in rodents.
Collapse
|
40
|
Current Transport Systems and Clinical Applications for Small Interfering RNA (siRNA) Drugs. Mol Diagn Ther 2019; 22:551-569. [PMID: 29926308 DOI: 10.1007/s40291-018-0338-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Small interfering RNAs (siRNAs) are an attractive new agent with potential as a therapeutic tool because of its ability to inhibit specific genes for many conditions, including viral infections and cancers. However, despite this potential, many challenges remain, including off-target effects, difficulties with delivery, immune responses, and toxicity. Traditional genetic vectors do not guarantee that siRNAs will silence genes in vivo. Rational design strategies, such as chemical modification, viral vectors, and non-viral vectors, including cationic liposomes, polymers, nanocarriers, and bioconjugated siRNAs, provide important opportunities to overcome these challenges. We summarize the results of research into vector delivery of siRNAs as a therapeutic agent from their design to clinical trials in ophthalmic diseases, cancers, respiratory diseases, and liver virus infections. Finally, we discuss the current state of siRNA delivery methods and the need for greater understanding of the requirements.
Collapse
|
41
|
Ramishetti S, Peer D. Engineering lymphocytes with RNAi. Adv Drug Deliv Rev 2019; 141:55-66. [PMID: 30529305 DOI: 10.1016/j.addr.2018.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/31/2018] [Accepted: 12/03/2018] [Indexed: 12/24/2022]
Abstract
Lymphocytes are the gatekeepers of the body's immune system and are involved in pathogenesis if their surveillance is stalled by inhibitory molecules or when they act as mediators for viral entry. Engineering lymphocytes in order to restore their functions is an unmet need in immunological disorders, cancer and in lymphotropic viral infections. Recently, the FDA approved several therapeutic antibodies for blocking inhibitory signals on T cells. This has revolutionized the field of solid tumor care, together with chimeric antigen receptor T cell (CAR-T) therapy that did the same for hematological malignancies. RNA interference (RNAi) is a promising approach where gene function can be inhibited in almost all types of cells. However, manipulation of genes in lymphocyte subsets are difficult due to their hard-to-transfect nature and in vivo targeting remains challenging as they are dispersed throughout the body. The ability of RNAi molecules to gain entry into cells is almost impossible without delivery strategy. Nanotechnology approaches are rapidly growing and their impact in the field of drug and gene delivery applications to transport payloads inside cells have been extensively studied. Here we discuss various technologies available for RNAi delivery to lymphocytes. We shed light on the importance of targeting molecules in order to target lymphocytes in vivo. In addition, we discuss recent developments of RNAi delivery to lymphocyte subsets, and detail the potential implication for the future of molecular medicine in leukocytes implicated diseases.
Collapse
|
42
|
Hesari A, Anoshiravani AA, Talebi S, Noruzi S, Mohammadi R, Salarinia R, Zare R, Ghasemi F. Knockdown of sal-like 4 expression by small interfering RNA induces apoptosis in breast cancer cells. J Cell Biochem 2018; 120:9392-9399. [PMID: 30520112 DOI: 10.1002/jcb.28214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 11/15/2018] [Indexed: 12/21/2022]
Abstract
Breast cancer is the most prevalent cancers worldwide and causes a significant amount of deaths annually. Spalt-like transcription factor 4 is known as a transcription factor, which has an important role in the proliferation of cancerous cells. Small interfering RNA (siRNA) is a short-chain molecule of 20 to 25 nucleotides that protrude on two sides of the 3', two nucleotides. In this study, using a specific sequence of siRNA against the sequence of this gene, its activity is investigated in the cell line of breast cancer. The breast cancer cells (MCF-7) were cultured and then, using a specific anti-sal-like 4 (SALL4) siRNA, their toxic doses were determined. Then, the gene is transfected into the cell. Proliferation and expression of the SALL4 and BCL-2 gene were measured using the real-time polymerase chain reaction method. The specific concentration of siRNA IC50 of the SALL4 gene was 40.35 nmole. Gene expression results indicated that the expression of the Bcl-2 gene in the siRNA group was significantly reduced ( P < 0.05). SiRNA can increase the apoptosis of breast cancer cells by reducing the gene expression of SALL4 gene and Bcl-2; it can be used as a novel targeted therapy. This strategy, in addition to increasing the specificity of the drug, also reduces the side effects when compared with conventional chemotherapy.
Collapse
Affiliation(s)
- Amireza Hesari
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | | | - Samaneh Talebi
- Division of Human Genetics, Immunology Research Center, Avicenna Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Somayye Noruzi
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Rezvan Mohammadi
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Reza Salarinia
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.,Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Reza Zare
- Student Research Committee, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Faezeh Ghasemi
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.,Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Next to Milad Tower, Tehran, Iran
| |
Collapse
|
43
|
Su L, Tian J, Sun J, Han N, Feng L, Yu B, Wang Y. Lentivirus-mediated siRNA knockdown of SPHK1 inhibits proliferation and tumorigenesis of neuroblastoma. Onco Targets Ther 2018; 11:7187-7196. [PMID: 30425511 PMCID: PMC6203087 DOI: 10.2147/ott.s180962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The overexpression of sphingosine kinase 1 (SPHK1) is responsible for the progress of many cancers. However, the role of SPHK1 in the development and progression of neuroblastoma (NB) remain largely unknown. Here in this study, we explored whether silencing SPHK1 by lentivirus-mediated siRNA could be employed as a potential therapeutic target for NB. MATERIALS AND METHODS Lentivirus was adopted to load SPHK1 siRNA. The results were obtained using RT-qPCR, Western blot, cell proliferation assay, transwell cell migration/invasion assays as well as in vivo xenograft tumor models in nude mice. RESULTS Our results demonstrated that SPHK1 mRNA was upregulated in SH-SY5Y and SK-N-SH cells as well as in human NB tissues. SPHK1 knockdown by siRNA resulted in impaired proliferation, increased apoptosis, as well as impaired migration and invasion of SH-SY5Y and SK-N-SH cells. In addition, the in vivo study suggested that SPHK1 knockdown significantly reduced the tumorigenesis of SH-SY5Y xenograft model. Furthermore, intratumorally administered lentivirus-SPHK1 siRNA could significantly inhibit tumor growth in an SH-SY5Y xenograft mice model. Intensive investigations on mechanism revealed that these effects were achieved through the deactivation of STAT3 pathways. CONCLUSION These data suggest that SPHK1 inhibition via downregulation of STAT3 pathways by lentivirus-mediated siRNA knockdown can significantly suppress NB progression, which could be a promising target for future gene therapy of NB.
Collapse
Affiliation(s)
- Lin Su
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Junyan Tian
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Jinsong Sun
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Nuan Han
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Lin Feng
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Baohua Yu
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| | - Yuepeng Wang
- Department of Pediatric Surgery, Affiliated Hospital of Jining Medical University, Jining City 272029, Shandong Province, People's Republic of China,
| |
Collapse
|
44
|
Liu Y, Song Z, Zheng N, Nagasaka K, Yin L, Cheng J. Systemic siRNA delivery to tumors by cell-penetrating α-helical polypeptide-based metastable nanoparticles. NANOSCALE 2018; 10:15339-15349. [PMID: 30070662 PMCID: PMC6734929 DOI: 10.1039/c8nr03976c] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Systemic, non-viral siRNA delivery for cancer treatment is mainly achieved via condensation by cationic materials (e.g., lipids and cationic polymers), which nevertheless, suffers from poor serum stability, non-specific tissue interaction, and unsatisfactory membrane activity against efficient in vivo gene knockdown. Here, we report the design of a metastable, cancer-targeting siRNA delivery system based on two functional polymers, PVBLG-8, a cationic, helical cell-penetrating polypeptide, and poly(l-glutamic acid) (PLG), an anionic random-coiled polypeptide. PVBLG-8 with rigid, linear structure showed weak siRNA condensation capability, and PLG with flexible chains was incorporated as a stabilizer which provided sufficient molecular entanglement with PVBLG-8 to encapsulate the siRNA within the polymeric network. The obtained PVBLG-8/siRNA/PLG nanoparticles (PSP NPs) with positive charges were sequentially coated with additional amount of PLG, which reversed the surface charge from positive to negative to yield the metastable PVBLG-8/siRNA/PLG@PLG (PSPP) NPs. The PSPP NPs featured desired serum stability during circulation to enhance tumor accumulation via the enhanced permeability and retention (EPR) effect. Upon acidification in the tumor extracellular microenvironment and intracellular endosomes, the partial protonation of PLG on PSPP NPs surface would lead to dissociation of PLG coating from NPs, exposure of the highly membrane-active PVBLG-8, and surface charge reversal from negative to positive, which subsequently promoted tumor penetration, selective cancer cell internalization, and efficient endolysosomal escape. When siRNA against epidermal growth factor receptor (EGFR) was encapsulated, the PSPP NPs showed excellent tumor penetration capability, tumor cell uptake level, EGFR silencing efficiency, and tumor growth inhibition efficacy in U-87 MG glioblastoma tumor spheroids in vitro and in xenograft tumor-bearing mice in vivo, outperforming the PSP NPs and several commercial reagents such as Lipofectamine 2000 and poly(l-lysine) (PLL). This study therefore demonstrates a facile and unique design approach of metastable and charge reversal NPs, which overcomes multiple biological barriers against systemic siRNA delivery toward anti-cancer treatment.
Collapse
Affiliation(s)
- Yang Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Illinois 61801, USA
| | - Ziyuan Song
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Illinois 61801, USA
| | - Nan Zheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Illinois 61801, USA
| | - Kenya Nagasaka
- School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Illinois 61801, USA
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Corresponding author: (J.C.); (L.Y.)
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Illinois 61801, USA
- Frederick Seitz Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Micro and Nanotechnology Lab, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Corresponding author: (J.C.); (L.Y.)
| |
Collapse
|
45
|
Dzmitruk V, Apartsin E, Ihnatsyeu-Kachan A, Abashkin V, Shcharbin D, Bryszewska M. Dendrimers Show Promise for siRNA and microRNA Therapeutics. Pharmaceutics 2018; 10:E126. [PMID: 30096839 PMCID: PMC6161126 DOI: 10.3390/pharmaceutics10030126] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/24/2022] Open
Abstract
The lack of an appropriate intracellular delivery system for therapeutic nucleic acids (TNAs) is a major problem in molecular biology, biotechnology, and medicine. A relatively new class of highly symmetrical hyperbranched polymers, called dendrimers, shows promise for transporting small TNAs into both cells and target tissues. Dendrimers have intrinsic advantages for this purpose: their physico-chemical and biological properties can be controlled during synthesis, and they are able to transport large numbers of TNA molecules that can specifically suppress the expression of single or multiple targeted genes. Numerous chemical modifications of dendrimers extend the biocompatibility of synthetic materials and allow targeted vectors to be designed for particular therapeutic purposes. This review summarizes the latest experimental data and trends in the medical application of various types of dendrimers and dendrimer-based nanoconstructions as delivery systems for short small interfering RNAs (siRNAs) and microRNAs at the cell and organism levels. It provides an overview of the structural features of dendrimers, indicating their advantages over other types of TNA transporters.
Collapse
Affiliation(s)
- Volha Dzmitruk
- Institute of Biophysics and Cell Engineering of NASB, 220072 Minsk, Belarus.
| | - Evgeny Apartsin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia.
| | - Aliaksei Ihnatsyeu-Kachan
- Institute of Biophysics and Cell Engineering of NASB, 220072 Minsk, Belarus.
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), 02972 Seoul, Korea.
| | - Viktar Abashkin
- Institute of Biophysics and Cell Engineering of NASB, 220072 Minsk, Belarus.
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of NASB, 220072 Minsk, Belarus.
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| |
Collapse
|
46
|
Kim GH, Won JE, Byeon Y, Kim MG, Wi TI, Lee JM, Park YY, Lee JW, Kang TH, Jung ID, Shin BC, Ahn HJ, Lee YJ, Sood AK, Han HD, Park YM. Selective delivery of PLXDC1 small interfering RNA to endothelial cells for anti-angiogenesis tumor therapy using CD44-targeted chitosan nanoparticles for epithelial ovarian cancer. Drug Deliv 2018; 25:1394-1402. [PMID: 29890852 PMCID: PMC6096458 DOI: 10.1080/10717544.2018.1480672] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Angiogenesis plays an essential role in the growth and metastasis of tumor cells, and the modulation of angiogenesis can be an effective approach for cancer therapy. We focused on silencing the angiogenic gene PLXDC1 as an important factor for anti-angiogenesis tumor therapy. Herein, we developed PLXDC1 small interfering siRNA (siRNA)-incorporated chitosan nanoparticle (CH-NP/siRNA) coated with hyaluronic acid (HA) to target the CD44 receptor on tumor endothelial cells. This study aimed to improve targeted delivery and enhance therapeutic efficacy for tumor anti-angiogenesis. The HA-CH-NP/siRNA was 200 ± 10 nm in size with a zeta potential of 26.4 mV. The loading efficiency of siRNA to the HA-CH-NP/siRNA was up to 60%. The selective binding of HA-CH-NP/siRNA to CD44-positive tumor endothelial cells increased by 2.1-fold compared with that of the CD44 nontargeted CH-NP/siRNA. PLXDC1 silencing by the HA-CH-NP/siRNA significantly inhibited tumor growth in A2780 tumor-bearing mice compared with that in the control group (p < .01), and mRNA expression of PLXDC1 was significantly reduced in the HA-CH-NP/siRNA-treated group. Furthermore, treatment with HA-CH-NP/siRNA resulted in significant inhibition of cell proliferation (p < .001), reduced microvessel density (p < .001), and increased cell apoptosis (p < .001). This study demonstrates that HA-CH-NP/siRNA is a highly selective delivery platform for siRNA, and has broad potential to be used in anti-angiogenesis tumor therapy.
Collapse
Affiliation(s)
- Ga Hee Kim
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| | - Ji Eun Won
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| | - Yeongseon Byeon
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| | - Min Gi Kim
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| | - Tae In Wi
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| | - Jae Myeong Lee
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| | - Yun-Yong Park
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Tae Heung Kang
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| | - In Duk Jung
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| | - Byung Cheol Shin
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Hyung Jun Ahn
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Young Joo Lee
- Department of Bioscience and Biotechnology, Sejong University, Kwang-Jin-Gu, Seoul, Republic of Korea
| | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hee Dong Han
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| | - Yeong-Min Park
- Department of Immunology School of Medicine, Konkuk University, Chungju, South Korea
| |
Collapse
|
47
|
Kim B, Pang HB, Kang J, Park JH, Ruoslahti E, Sailor MJ. Immunogene therapy with fusogenic nanoparticles modulates macrophage response to Staphylococcus aureus. Nat Commun 2018; 9:1969. [PMID: 29773788 PMCID: PMC5958120 DOI: 10.1038/s41467-018-04390-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 04/25/2018] [Indexed: 01/06/2023] Open
Abstract
The incidence of adverse effects and pathogen resistance encountered with small molecule antibiotics is increasing. As such, there is mounting focus on immunogene therapy to augment the immune system’s response to infection and accelerate healing. A major obstacle to in vivo gene delivery is that the primary uptake pathway, cellular endocytosis, results in extracellular excretion and lysosomal degradation of genetic material. Here we show a nanosystem that bypasses endocytosis and achieves potent gene knockdown efficacy. Porous silicon nanoparticles containing an outer sheath of homing peptides and fusogenic liposome selectively target macrophages and directly introduce an oligonucleotide payload into the cytosol. Highly effective knockdown of the proinflammatory macrophage marker IRF5 enhances the clearance capability of macrophages and improves survival in a mouse model of Staphyloccocus aureus pneumonia. In the context of increasing bacterial antibiotic-resistance, gene therapy that targets the immune system to clear infection is a major goal. Here the authors show a silicon based nanosystem that modulates the macrophage response in an in vivo model of Staphylococcal pneumonia.
Collapse
Affiliation(s)
- Byungji Kim
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Hong-Bo Pang
- Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA.,Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Jinyoung Kang
- Department of Nanoengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Ji-Ho Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Erkki Ruoslahti
- Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, 92037, USA.,Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, 93106-9610, USA
| | - Michael J Sailor
- Materials Science and Engineering Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA. .,Department of Nanoengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA. .,Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA.
| |
Collapse
|
48
|
Xie Y, Wang Y, Li J, Hang Y, Oupický D. Promise of chemokine network-targeted nanoparticles in combination nucleic acid therapies of metastatic cancer. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1528. [PMID: 29700990 DOI: 10.1002/wnan.1528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/28/2018] [Accepted: 04/01/2018] [Indexed: 01/10/2023]
Abstract
Chemokines and chemokine receptors play key roles in cancer progression and metastasis. Although multiple chemokines and chemokine receptors have been investigated, inhibition of CXCR4 emerged as one of the most promising approaches in combination cancer therapy, especially when focused on the metastatic disease. Small RNA molecules, such as small interfering RNA (siRNA) and microRNA (miRNA), represent new class of therapeutics for cancer treatment through RNA interference-mediated gene silencing. However, the clinical applicability of siRNA and miRNA is severely limited by the lack of effective delivery systems. There is a significant therapeutic potential for CXCR4-targeted nanomedicines in combination with the delivery of siRNA and miRNA in cancer. Recently developed CXCR4-targeted polymeric drugs and nanomedicines, including cyclam- and chloroquine-based polymeric CXCR4 antagonists are introduced here and their ability to deliver functional siRNA and miRNA is discussed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Ying Xie
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yazhe Wang
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jing Li
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yu Hang
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - David Oupický
- Department of Pharmaceutical Sciences, Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
49
|
Leimena C, Qiu H. Non-Coding RNA in the Pathogenesis, Progression and Treatment of Hypertension. Int J Mol Sci 2018; 19:E927. [PMID: 29561765 PMCID: PMC5979335 DOI: 10.3390/ijms19040927] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 12/11/2022] Open
Abstract
Hypertension is a complex, multifactorial disease that involves the coexistence of multiple risk factors, environmental factors and physiological systems. The complexities extend to the treatment and management of hypertension, which are still the pursuit of many researchers. In the last two decades, various genes have emerged as possible biomarkers and have become the target for investigations of specialized drug design based on its risk factors and the primary cause. Owing to the growing technology of microarrays and next-generation sequencing, the non-protein-coding RNAs (ncRNAs) have increasingly gained attention, and their status of redundancy has flipped to importance in normal cellular processes, as well as in disease progression. The ncRNA molecules make up a significant portion of the human genome, and their role in diseases continues to be uncovered. Specifically, the cellular role of these ncRNAs has played a part in the pathogenesis of hypertension and its progression to heart failure. This review explores the function of the ncRNAs, their types and biology, the current update of their association with hypertension pathology and the potential new therapeutic regime for hypertension.
Collapse
Affiliation(s)
- Christiana Leimena
- Department of Basic Sciences, Physiological Division, School of Medicine, Loma Linda University, Loma Linda, CA 92324, USA.
| | - Hongyu Qiu
- Department of Basic Sciences, Physiological Division, School of Medicine, Loma Linda University, Loma Linda, CA 92324, USA.
| |
Collapse
|
50
|
Zheng M, Tao W, Zou Y, Farokhzad OC, Shi B. Nanotechnology-Based Strategies for siRNA Brain Delivery for Disease Therapy. Trends Biotechnol 2018; 36:562-575. [PMID: 29422412 DOI: 10.1016/j.tibtech.2018.01.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/10/2018] [Accepted: 01/11/2018] [Indexed: 02/08/2023]
Abstract
Small interfering RNA (siRNA)-based gene silencing technology has demonstrated significant potential for treating brain-associated diseases. However, effective and safe systemic delivery of siRNA into the brain remains challenging because of biological barriers such as enzymatic degradation, short circulation lifetime, the blood-brain barrier (BBB), insufficient tissue penetration, cell endocytosis, and cytosolic transport. Nanotechnology offers intriguing potential for addressing these challenges in siRNA brain delivery in conjunction with chemical and biological modification strategies. In this review, we outline the challenges of systemic delivery of siRNA-based therapy for brain diseases, highlight recent advances in the development and engineering of siRNA nanomedicines for various brain diseases, and discuss our perspectives on this exciting research field for siRNA-based therapy towards more effective brain disease therapy.
Collapse
Affiliation(s)
- Meng Zheng
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yan Zou
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bingyang Shi
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|