1
|
Kreofsky NW, Roy P, Reineke TM. pH-Responsive Micelles Containing Quinine Functionalities Enhance Intracellular Gene Delivery and Expression. Bioconjug Chem 2024. [PMID: 39467734 DOI: 10.1021/acs.bioconjchem.4c00326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Quinine is a promising building block for creating polymer carriers for intracellular nucleic acid delivery. This is due to its ability to bind to genetic material through intercalation and electrostatic interactions and the balance of hydrophobicity and hydrophilicity dependent on the pH/charge state. Yet, studies utilizing cinchona alkaloid natural products in gene delivery are limited. Herein, we present the incorporation of a quinine functionalized monomer (Q) into block polymer architectures to form self-assembled micelles for highly efficient gene delivery. Q was incorporated into the core and/or the shell of the micelles to introduce the unique advantages of quinine to the system. We found that incorporation of Q into the core of the micelle resulted in acid-induced disassembly of the micelle and a boost in transfection efficiency by promoting endosomal escape. This effect was especially evident in the cancerous cell line, A549, which has a more acidic intracellular environment. Incorporation of Q into the shell of the micelles resulted in intercalative binding to the genetic payload as well as larger micelle-DNA complexes (micelleplexes) from the hydrophobicity of Q in the shell. These factors enable the micelleplexes to be more resistant to serum and have more persistent protein expression post-transfection. Overall, this study is the first to demonstrate the benefits of including quinine functionalities into self-assembled micelles for highly efficient gene delivery and presents a platform for inclusion of other natural products with similar properties into micellar systems.
Collapse
Affiliation(s)
- Nicholas W Kreofsky
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Punarbasu Roy
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
2
|
Xing Y, Meng B, Chen Q. Cyclodextrin-Containing Drug Delivery Systems and Their Applications in Neurodegenerative Disorders. Int J Mol Sci 2024; 25:10834. [PMID: 39409162 PMCID: PMC11477047 DOI: 10.3390/ijms251910834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/20/2024] Open
Abstract
Cyclodextrins (CDs) are ubiquitous excipients, constituted of cyclic glucopyranose units, and possess a unique dual nature, that of a hydrophobic interior and a hydrophilic exterior. This enables their interaction with lipid-affinitive compounds and hydrophilic compounds, thereby augmenting their application in pharmaceutical formulations as agents for improving solubility, as well as fundamental elements of advanced drug delivery systems. Additionally, CDs, upon suitable modification, can strategically participate in the interaction with cellular components and physical barriers, such as the blood-brain barrier, where their intricate and multifunctional engagement leads to various biological impacts. This review consolidates the crucial features of CDs and their derivatives, and summarizes the applications of them as drug delivery systems in neurodegenerative disorders, emphasizing their notable potentials.
Collapse
Affiliation(s)
- Yuan Xing
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China; (Y.X.); (B.M.)
| | - Bohan Meng
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China; (Y.X.); (B.M.)
| | - Qi Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, China; (Y.X.); (B.M.)
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
3
|
Dastgerdi NK, Dastgerdi NK, Bayraktutan H, Costabile G, Atyabi F, Dinarvand R, Longobardi G, Alexander C, Conte C. Enhancing siRNA cancer therapy: Multifaceted strategies with lipid and polymer-based carrier systems. Int J Pharm 2024; 663:124545. [PMID: 39098747 DOI: 10.1016/j.ijpharm.2024.124545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Cancers are increasing in prevalence and many challenges remain for their treatment, such as chemoresistance and toxicity. In this context, siRNA-based therapeutics have many potential advantages for cancer therapies as a result of their ability to reduce or prevent expression of specific cancer-related genes. However, the direct delivery of naked siRNA is hindered by issues like enzymatic degradation, insufficient cellular uptake, and poor pharmacokinetics. Hence, the discovery of a safe and efficient delivery vehicle is essential. This review explores various lipid and polymer-based delivery systems for siRNA in cancer treatment. Both polymers and lipids have garnered considerable attention as carriers for siRNA delivery. While all of these systems protect siRNA and enhance transfection efficacy, each exhibits its unique strengths. Lipid-based delivery systems, for instance, demonstrate high entrapment efficacy and utilize cost-effective materials. Conversely, polymeric-based delivery systems offer advantages through chemical modifications. Nonetheless, certain drawbacks still limit their usage. To address these limitations, combining different materials in formulations (lipid, polymer, or targeting agent) could enhance pharmaceutical properties, boost transfection efficacy, and reduce side effects. Furthermore, co-delivery of siRNA with other therapeutic agents presents a promising strategy to overcome cancer resistance. Lipid-based delivery systems have been demonstrated to encapsulate many therapeutic agents and with high efficiency, but most are limited in terms of the functionalities they display. In contrast, polymeric-based delivery systems can be chemically modified by a wide variety of routes to include multiple components, such as release or targeting elements, from the same materials backbone. Accordingly, by incorporating multiple materials such as lipids, polymers, and/or targeting agents in RNA formulations it is possible to improve the pharmaceutical properties and therapeutic efficacy while reducing side effects. This review focuses on strategies to improve siRNA cancer treatments and discusses future prospects in this important field.
Collapse
Affiliation(s)
- Nazgol Karimi Dastgerdi
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK; Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Karimi Dastgerdi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hulya Bayraktutan
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | | | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran.
| | | | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | - Claudia Conte
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy.
| |
Collapse
|
4
|
Oliveira IS, Silva SG, Gomes AC, Real Oliveira MECD, Vale MLCD, Marques EF. Cationic Serine-Based Gemini Surfactant:Monoolein Aggregates as Viable and Efficacious Agents for DNA Complexation and Compaction: A Cytotoxicity and Physicochemical Assessment. J Funct Biomater 2024; 15:224. [PMID: 39194661 DOI: 10.3390/jfb15080224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
Cationic gemini surfactants have emerged as potential gene delivery agents as they can co-assemble with DNA due to a strong electrostatic association. Commonly, DNA complexation is enhanced by the inclusion of a helper lipid (HL), which also plays a key role in transfection efficiency. The formation of lipoplexes, used as non-viral vectors for transfection, through electrostatic and hydrophobic interactions is affected by various physicochemical parameters, such as cationic surfactant:HL molar ratio, (+/-) charge ratio, and the morphological structure of the lipoplexes. Herein, we investigated the DNA complexation ability of mixtures of serine-based gemini surfactants, (nSer)2N5, and monoolein (MO) as a helper lipid. The micelle-forming serine surfactants contain long lipophilic chains (12 to 18 C atoms) and a five CH2 spacer, both linked to the nitrogen atoms of the serine residues by amine linkages. The (nSer)2N5:MO aggregates are non-cytotoxic up to 35-90 µM, depending on surfactant and surfactant/MO mixing ratio, and in general, higher MO content and longer surfactant chain length tend to promote higher cell viability. All systems efficaciously complex DNA, but the (18Ser)2N5:MO one clearly stands as the best-performing one. Incorporating MO into the serine surfactant system affects the morphology and size distribution of the formed mixed aggregates. In the low concentration regime, gemini-MO systems aggregate in the form of vesicles, while at high concentrations the formation of a lamellar liquid crystalline phase is observed. This suggests that lipoplexes might share a similar bilayer-based structure.
Collapse
Affiliation(s)
- Isabel S Oliveira
- CIQUP (Centro de Investigação em Química da Universidade do Porto), IMS (Institute of Molecular Sciences), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Sandra G Silva
- LAQV-REQUIMTE (Laboratório Associado para a Química Verde-Rede Química e Tecnologia), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Andreia C Gomes
- CBMA (Centro de Biologia Molecular e Ambiental), Departamento de Biologia, Campus de Gualtar, Universidade do Minho, 4710-057 Braga, Portugal
| | - M Elisabete C D Real Oliveira
- CFUM (Center of Physics), Departamento de Física, Universidade do Minho, Campos de Gualtar, 4710-057 Braga, Portugal
| | - M Luísa C do Vale
- LAQV-REQUIMTE (Laboratório Associado para a Química Verde-Rede Química e Tecnologia), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Eduardo F Marques
- CIQUP (Centro de Investigação em Química da Universidade do Porto), IMS (Institute of Molecular Sciences), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| |
Collapse
|
5
|
Jogdeo CM, Siddhanta K, Das A, Ding L, Panja S, Kumari N, Oupický D. Beyond Lipids: Exploring Advances in Polymeric Gene Delivery in the Lipid Nanoparticles Era. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404608. [PMID: 38842816 PMCID: PMC11384239 DOI: 10.1002/adma.202404608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/23/2024] [Indexed: 06/07/2024]
Abstract
The recent success of gene therapy during the COVID-19 pandemic has underscored the importance of effective and safe delivery systems. Complementing lipid-based delivery systems, polymers present a promising alternative for gene delivery. Significant advances have been made in the recent past, with multiple clinical trials progressing beyond phase I and several companies actively working on polymeric delivery systems which provides assurance that polymeric carriers can soon achieve clinical translation. The massive advantage of structural tunability and vast chemical space of polymers is being actively leveraged to mitigate shortcomings of traditional polycationic polymers and improve the translatability of delivery systems. Tailored polymeric approaches for diverse nucleic acids and for specific subcellular targets are now being designed to improve therapeutic efficacy. This review describes the recent advances in polymer design for improved gene delivery by polyplexes and covalent polymer-nucleic acid conjugates. The review also offers a brief note on novel computational techniques for improved polymer design. The review concludes with an overview of the current state of polymeric gene therapies in the clinic as well as future directions on their translation to the clinic.
Collapse
Affiliation(s)
- Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ashish Das
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Neha Kumari
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
6
|
Chen J, Li F, Zhao B, Gu J, Brejcha NM, Bartoli M, Zhang W, Zhou Y, Fu S, Domena JB, Zafar A, Zhang F, Tagliaferro A, Verde F, Zhang F, Zhang Y, Leblanc RM. Gene Transfection Efficiency Improvement with Lipid Conjugated Cationic Carbon Dots. ACS APPLIED MATERIALS & INTERFACES 2024; 16:27087-27101. [PMID: 38752799 DOI: 10.1021/acsami.4c02614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
An ideal vehicle with a high transfection efficiency is crucial for gene delivery. In this study, a type of cationic carbon dot (CCD) known as APCDs were first prepared with arginine (Arg) and pentaethylenehexamine (PEHA) as precursors and conjugated with oleic acid (OA) for gene delivery. By tuning the mass ratio of APCDs to OA, APCDs-OA conjugates, namely, APCDs-0.5OA, APCDs-1.0OA, and APCDs-1.5OA were synthesized. All three amphiphilic APCDs-OA conjugates show high affinity to DNA through electrostatic interactions. APCDs-0.5OA exhibit strong binding with small interfering RNA (siRNA). After being internalized by Human Embryonic Kidney (HEK 293) and osteosarcoma (U2OS) cells, they could distribute in both the cytoplasm and the nucleus. With APCDs-OA conjugates as gene delivery vehicles, plasmid DNA (pDNA) that encodes the gene for the green fluorescence protein (GFP) can be successfully delivered in both HEK 293 and U2OS cells. The GFP expression levels mediated by APCDs-0.5OA and APCDs-1.0OA are ten times greater than that of PEI in HEK 293 cells. Furthermore, APCDs-0.5OA show prominent siRNA transfection efficiency, which is proven by the significantly downregulated expression of FANCA and FANCD2 proteins upon delivery of FANCA siRNA and FANCD2 siRNA into U2OS cells. In conclusion, our work demonstrates that conjugation of CCDs with a lipid structure such as OA significantly improves the gene transfection efficiency, providing a new idea about the designation of nonviral carriers in gene delivery systems.
Collapse
Affiliation(s)
- Jiuyan Chen
- Department of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| | - Fang Li
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Bowen Zhao
- Department of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| | - Jun Gu
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Nicholas Michael Brejcha
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Mattia Bartoli
- Department of Applied Science and Technology, Politecnico di Torino, Torino 10129, Italy
| | - Wei Zhang
- Department of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| | - Yiqun Zhou
- Department of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| | - Shiwei Fu
- Department of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| | - Justin B Domena
- Department of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| | - Alyan Zafar
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Fuwu Zhang
- Department of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| | - Alberto Tagliaferro
- Department of Applied Science and Technology, Politecnico di Torino, Torino 10129, Italy
| | - Fulvia Verde
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Fangliang Zhang
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Yanbin Zhang
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, Coral Gables, Florida 33146, United States
| |
Collapse
|
7
|
Zhang Y, Zhang M, Song H, Dai Q, Liu C. Tumor Microenvironment-Responsive Polymer-Based RNA Delivery Systems for Cancer Treatment. SMALL METHODS 2024:e2400278. [PMID: 38803312 DOI: 10.1002/smtd.202400278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/30/2024] [Indexed: 05/29/2024]
Abstract
Ribonucleic acid (RNA) therapeutics offer a broad prospect in cancer treatment. However, their successful application requires overcoming various physiological barriers to effectively deliver RNAs to the target sites. Currently, a number of RNA delivery systems based on polymeric nanoparticles are developed to overcome these barriers in RNA delivery. This work provides an overview of the existing RNA therapeutics for cancer gene therapy, and particularly summarizes those that are entering the clinical phase. This work then discusses the core features and latest research developments of tumor microenvironment-responsive polymer-based RNA delivery carriers which are designed based on the pathological characteristics of the tumor microenvironment. Finally, this work also proposes opportunities for the transformation of RNA therapies into cancer immunotherapy methods in clinical applications.
Collapse
Affiliation(s)
- Yahan Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ming Zhang
- Department of Pathology, Peking University International Hospital, Beijing, 102206, China
| | - Haiqin Song
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Qiong Dai
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
8
|
Feng Y, Wei C, Gu Y, Zhang H, Liu L, Chen Y, Zhao T. pH-sensitive cationic nanoparticles for endosomal cell-free DNA scavenging against acute inflammation. J Control Release 2024; 369:88-100. [PMID: 38471640 DOI: 10.1016/j.jconrel.2024.03.003] [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: 12/29/2023] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
Cell-free DNA (cfDNA) released from dead cells could be a player in some autoimmune disorders by activating Toll-like receptor 9 (TLR9) and inducing proinflammatory cytokines. Cationic nanoparticles (cNPs) address cfDNA clearance, yet challenges persist, including toxicity, low specificity and ineffectiveness against endocytosed cfDNA. This study introduced pH-sensitive cNPs, reducing off-target effects and binding cfDNA at inflammatory sites. This unique approach inhibits the TLR9 pathway, offering a novel strategy for inflammation modulation. Synthesized cNPs, with distinct cationic moieties, exhibit varied pKa values, enhancing cfDNA binding. Comprehensive studies elucidate the mechanism, demonstrating minimal extracellular binding, enhanced endosomal DNA binding, and optimal tumor necrosis factor-α suppression. In a traumatic brain injury mice model, pH-sensitive cNPs effectively suppress inflammatory cytokines, highlighting their potential in acute inflammation regulation.
Collapse
Affiliation(s)
- Yilin Feng
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Cong Wei
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Yanrong Gu
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Hong Zhang
- Department of Biomedical Engineering, Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Jinan University, Guangzhou 510632, China
| | - Lixin Liu
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China; Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Yongming Chen
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China; Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China; Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; College of Chemistry and Molecular Science, Henan University, Zhengzhou, China.
| | - Tianyu Zhao
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
9
|
Imani KBC, Dodda JM, Yoon J, Torres FG, Imran AB, Deen GR, Al‐Ansari R. Seamless Integration of Conducting Hydrogels in Daily Life: From Preparation to Wearable Application. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306784. [PMID: 38240470 PMCID: PMC10987148 DOI: 10.1002/advs.202306784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/12/2023] [Indexed: 04/04/2024]
Abstract
Conductive hydrogels (CHs) have received significant attention for use in wearable devices because they retain their softness and flexibility while maintaining high conductivity. CHs are well suited for applications in skin-contact electronics and biomedical devices owing to their high biocompatibility and conformality. Although highly conductive hydrogels for smart wearable devices are extensively researched, a detailed summary of the outstanding results of CHs is required for a comprehensive understanding. In this review, the recent progress in the preparation and fabrication of CHs is summarized for smart wearable devices. Improvements in the mechanical, electrical, and functional properties of high-performance wearable devices are also discussed. Furthermore, recent examples of innovative and highly functional devices based on CHs that can be seamlessly integrated into daily lives are reviewed.
Collapse
Affiliation(s)
- Kusuma Betha Cahaya Imani
- Graduate Department of Chemical MaterialsInstitute for Plastic Information and Energy MaterialsSustainable Utilization of Photovoltaic Energy Research CenterPusan National UniversityBusan46241Republic of Korea
| | - Jagan Mohan Dodda
- New Technologies – Research Centre (NTC)University of West Bohemia, Univerzitní 8Pilsen301 00Czech Republic
| | - Jinhwan Yoon
- Graduate Department of Chemical MaterialsInstitute for Plastic Information and Energy MaterialsSustainable Utilization of Photovoltaic Energy Research CenterPusan National UniversityBusan46241Republic of Korea
| | - Fernando G. Torres
- Department of Mechanical EngineeringPontificia Universidad Catolica del Peru. Av. Universitaria 1801Lima15088Peru
| | - Abu Bin Imran
- Department of ChemistryBangladesh University of Engineering and TechnologyDhaka1000Bangladesh
| | - G. Roshan Deen
- Materials for Medicine Research GroupSchool of MedicineThe Royal College of Surgeons in Ireland (RCSI)Medical University of BahrainBusaiteen15503Kingdom of Bahrain
| | - Renad Al‐Ansari
- Materials for Medicine Research GroupSchool of MedicineThe Royal College of Surgeons in Ireland (RCSI)Medical University of BahrainBusaiteen15503Kingdom of Bahrain
| |
Collapse
|
10
|
Mahdieh A, Motasadizadeh H, Maghsoudian S, Sabzevari A, Khalili F, Yeganeh H, Nyström B. Novel polyurethane-based ionene nanoparticles electrostatically stabilized with hyaluronic acid for effective gene therapy. Colloids Surf B Biointerfaces 2024; 236:113802. [PMID: 38382225 DOI: 10.1016/j.colsurfb.2024.113802] [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: 10/18/2023] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
Gene therapy is considered to be a valuable strategy for effective cancer treatment. However, the development of effective delivery systems that can specifically deliver gene materials, such as siRNA to tumor tissues plays a critical role in cancer therapy. In the present study, we have developed a novel complex that is based on an electrostatic interaction between cationic polyurethane ionene (CPUI) nanoparticles and an anti-signal transducer and activator of transcription 3 (STAT3) siRNA. For active targeting, hyaluronic acid (HA) was used to coat the complexes, which significantly reduced the cytotoxicity of the blank nanocarriers while demonstrating high transport efficiency of the siRNA via the CD44-mediated endocytosis pathway in MCF-7 breast cancer cells. The targeted nanocarriers (HA/CPUI/siRNA) showed significantly higher cellular internalization in flow cytometry and confocal microscopy compared with the non-targeted system (CPUI/siRNA). In addition, the incorporation of HA on the surface of the complexes resulted in significantly greater suppression of the STAT3 gene compared to the corresponding non-targeted formulation. Whole-body fluorescence images showed more significant tumor accumulation of the targeted nanocarriers in 4T1 breast tumor-bearing mice. Therefore, HA/CPUI/siRNA nanocarriers are an interesting option for the siRNA-targeted treatment of breast cancer cells.
Collapse
Affiliation(s)
- Athar Mahdieh
- Department of Pharmacy, Section for Pharmaceutics and Social Pharmacy, University of Oslo, Oslo, Norway
| | - Hamidreza Motasadizadeh
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samane Maghsoudian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Sabzevari
- Polymer Faculty, Biomedical Engineering Department, Meybod University, Meybod, Yazd, Iran; Polymer Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Fereshte Khalili
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Yeganeh
- Iran Polymer and Petrochemical Institute, Tehran, Iran
| | - Bo Nyström
- Department of Chemistry, University of Oslo, Oslo, Norway.
| |
Collapse
|
11
|
Guo S, Wang J, Wang Q, Wang J, Qin S, Li W. Advances in peptide-based drug delivery systems. Heliyon 2024; 10:e26009. [PMID: 38404797 PMCID: PMC10884816 DOI: 10.1016/j.heliyon.2024.e26009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 02/27/2024] Open
Abstract
Drug delivery systems (DDSs) are designed to deliver drugs to their specific targets to minimize their toxic effects and improve their susceptibility to clearance during targeted transport. Peptides have high affinity, low immunogenicity, simple amino acid composition, and adjustable molecular size; therefore, most peptides can be coupled to drugs via linkers to form peptide-drug conjugates (PDCs) and act as active pro-drugs. PDCs are widely thought to be promising DDSs, given their ability to improve drug bio-compatibility and physiological stability. Peptide-based DDSs are often used to deliver therapeutic substances such as anti-cancer drugs and nucleic acid-based drugs, which not only slow the degradation rate of drugs in vivo but also ensure the drug concentration at the targeted site and prolong the half-life of drugs in vivo. This article provides an profile of the advancements and future development in functional peptide-based DDSs both domestically and internationally in recent years, in the expectation of achieving targeted drug delivery incorporating functional peptides and taking full advantage of synergistic effects.
Collapse
Affiliation(s)
- Sijie Guo
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Jing Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Qi Wang
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Jinxin Wang
- College of Life Sciences, Yantai University, Yantai, 264005, China
| | - Song Qin
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Wenjun Li
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| |
Collapse
|
12
|
Leer K, Reichel LS, Kimmig J, Richter F, Hoeppener S, Brendel JC, Zechel S, Schubert US, Traeger A. Optimization of Mixed Micelles Based on Oppositely Charged Block Copolymers by Machine Learning for Application in Gene Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306116. [PMID: 37794626 DOI: 10.1002/smll.202306116] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Indexed: 10/06/2023]
Abstract
The COVID-19 mRNA vaccines represent a milestone in developing non-viral gene carriers, and their success highlights the crucial need for continued research in this field to address further challenges. Polymer-based delivery systems are particularly promising due to their versatile chemical structure and convenient adaptability, but struggle with the toxicity-efficiency dilemma. Introducing anionic, hydrophilic, or "stealth" functionalities represents a promising approach to overcome this dilemma in gene delivery. Here, two sets of diblock terpolymers are created comprising hydrophobic poly(n-butyl acrylate) (PnBA), a copolymer segment made of hydrophilic 4-acryloylmorpholine (NAM), and either the cationic 3-guanidinopropyl acrylamide (GPAm) or the 2-carboxyethyl acrylamide (CEAm), which is negatively charged at neutral conditions. These oppositely charged sets of diblocks are co-assembled in different ratios to form mixed micelles. Since this experimental design enables countless mixing possibilities, a machine learning approach is applied to identify an optimal GPAm/CEAm ratio for achieving high transfection efficiency and cell viability with little resource expenses. After two runs, an optimal ratio to overcome the toxicity-efficiency dilemma is identified. The results highlight the remarkable potential of integrating machine learning into polymer chemistry to effectively tackle the enormous number of conceivable combinations for identifying novel and powerful gene transporters.
Collapse
Affiliation(s)
- Katharina Leer
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Liên S Reichel
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Julian Kimmig
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Stefan Zechel
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| |
Collapse
|
13
|
Leer K, Reichel LS, Wilhelmi M, Brendel JC, Traeger A. Tailoring Gene Transfer Efficacy through the Arrangement of Cationic and Anionic Blocks in Triblock Copolymer Micelles. ACS Macro Lett 2024:158-165. [PMID: 38230657 PMCID: PMC10883036 DOI: 10.1021/acsmacrolett.3c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
The arrangement of charged segments in triblock copolymer micelles affects the gene delivery potential of polymeric micelles and can increase the level of gene expression when an anionic segment is incorporated in the outer shell. Triblock copolymers were synthesized by RAFT polymerzation with narrow molar mass distributions and assembled into micelles with a hydrophobic core from poly(n-butyl acrylate). The ionic shell contained either (i) an anionic segment followed by a cationic segment (HAC micelles) or (ii) a cationic block followed by an anionic block (HCA micelles). The pH-responsive anionic block contained 2-carboxyethyl acrylamide (CEAm), while the cationic block comprised 3-guanidinopropyl acrylamide (GPAm). Increasing the molar content of CEAm in HAC and HCA micelles from 6 to 13 mol % improved cytocompatibility and the endosomal escape property, while the HCA micelle with the highest mol % of anionic charges in the outer shell exhibited the highest gene expression. It became evident that improved membrane interaction of the best performing HCA micelle contributed to achieving high gene expression.
Collapse
Affiliation(s)
- Katharina Leer
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Liên S Reichel
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Mara Wilhelmi
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter, Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry, Friedrich Schiller University Jena, Humboldtstrasse 10, 07743 Jena, Germany
- Jena Center for Soft Matter, Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
| |
Collapse
|
14
|
Lee Y, Ha J, Kim M, Kang S, Kang M, Lee M. Antisense-oligonucleotide co-micelles with tumor targeting peptides elicit therapeutic effects by inhibiting microRNA-21 in the glioblastoma animal models. J Adv Res 2023; 53:249-260. [PMID: 36632887 PMCID: PMC10658310 DOI: 10.1016/j.jare.2023.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/14/2022] [Accepted: 01/07/2023] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION miRNA-21 (miR-21) is highly expressed in glioblastoma, facilitating tumor growth by blocking the expression of apoptosis-related genes. Therefore, an antisense microRNA oligonucleotide (AMO) against miR-21 was suggested as a therapeutic nucleic acid for glioblastoma. OBJECTIVES AMO21 co-micelles were developed with tumor-targeting T7 peptides as an AMO21 delivery system by intranasal administration. METHODS Cholesterol-conjugated AMO21 (AMO21c) was mixed with cholesterol-conjugated T7 peptides (T7c) to produce tumor-targeted co-micelles. Physical characterization was performed by dynamic light scattering, gel retardation assay, scanning electron microscope and heparin competition assay. In vitro transfection efficiency to C6 glioblastoma cells was measured by flow cytometry. The AMO21c/T7c co-micelles were administered by intranasal instillation into the brain of intracranial glioblastoma rat models. Scrambled T7 (scrT7) and scrambled AMO21c (scrAMO21c) were used as a negative control. The therapeutic effects of the AMO21c/T7c co-micelles were evaluated by real time RT-PCR, immunohistochemistry, TUNEL assay, and Nissl staining. RESULTS The formation of the AMO21c/T7c co-micelles was confirmed in gel retardation and heparin competition assays. The highest delivery efficiency in vitro was achieved at a 1:10 wt ratio of AMO21c/T7c. The AMO21c/T7c co-micelles had higher delivery efficiency into C6 glioblastoma cells than naked AMO21c or AMO21c/lipofectamine complexes. After intranasal administration into the intracranial glioblastoma models, the delivery efficiency of the co-micelles into the brain was also higher than those of naked AMO21c and AMO21c/scrambled T7c. Thanks to their enhanced delivery efficiency, the AMO21c/T7c co-micelles downregulated miR-21, inducing the production of the pro-apoptotic phosphatase and tensin homolog (PTEN) and programmed cell death 4 (PDCD4) proteins in the tumor tissues. The tumor size was reduced by the AMO21c/T7c co-micelles more effectively than naked AMO21c, AMO21c/lipofectamine, or scrAMO21c/T7c treatment. CONCLUSION The results suggest that the co-micelles of AMO21c and T7c may be an efficient delivery system into a brain tumor through intranasal administration.
Collapse
Affiliation(s)
- Youngki Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
| | - Junkyu Ha
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
| | - Minkyung Kim
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
| | - Subin Kang
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
| | - Minji Kang
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea.
| |
Collapse
|
15
|
Grimme CJ, Hanson MG, Reineke TM. Enhanced ASO-Mediated Gene Silencing with Lipophilic pH-Responsive Micelles. Bioconjug Chem 2023. [PMID: 37384839 DOI: 10.1021/acs.bioconjchem.3c00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Herein, we examine the ASO-mediated gene silencing efficiency of pH-responsive micelles, by incorporating 2-(diisopropylamino)ethyl methacrylate (DIP) into the micelle core and comparing physical and biological properties with non-pH-responsive micelles. Additionally, the lipophilic effect of the micelle cores was examined in both types of micelles. Varying lipophilicity was achieved by varying alkyl monomer chain lengths─butyl (4), lauryl (12), and stearyl (18) methacrylate. Each of the micelles formed within our family offered the added benefit of well-defined and uniform templates for loading antisense oligonucleotide (ASO) payloads. Overall, the micelles followed previously established trends of outperforming their linear polymer (nonmicelle) analogs and ASO only control. More specifically, the highest performing micelles were the pH-responsive micelles with longer alkyl chains or higher lipophilicity─D-DIP+LMA and D-DIP+SMA (∼90% silencing). These two micelles demonstrated silencing efficiencies similar to Jet-PEI and Lipofectamine 2000 and caused lower toxicity than Lipofectamine 2000. The shortest alkyl chain pH-responsive micelle, D-DIP+BMA (64%), displayed strong gene silencing similar to that about that of its non-pH-responsive micelle, D-BMA (68%), and the pH-responsive micelle without an alkyl chain incorporated, D-DIP (59%). This work illuminates a minimum alkyl chain length dependence to allow gene silencing within our micelle family. However, including only longer alkyl chains into the micelle core without the pH-responsive unit DIP had a hindering effect, thus demonstrating the requirement of the DIP unit when including longer alkyl chain lengths. This work demonstrates the exemplary gene silencing efficiencies of polymeric micelles and uncovers the relationship between pH responsiveness and performance with lipophilic polymer micelles for enhancing ASO-mediated gene silencing.
Collapse
Affiliation(s)
- Christian J Grimme
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Avenue SE, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
16
|
Zhang YQ, Guo RR, Chen YH, Li TC, Du WZ, Xiang RW, Guan JB, Li YP, Huang YY, Yu ZQ, Cai Y, Zhang P, Ling GX. Ionizable drug delivery systems for efficient and selective gene therapy. Mil Med Res 2023; 10:9. [PMID: 36843103 PMCID: PMC9968649 DOI: 10.1186/s40779-023-00445-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 02/01/2023] [Indexed: 02/28/2023] Open
Abstract
Gene therapy has shown great potential to treat various diseases by repairing the abnormal gene function. However, a great challenge in bringing the nucleic acid formulations to the market is the safe and effective delivery to the specific tissues and cells. To be excited, the development of ionizable drug delivery systems (IDDSs) has promoted a great breakthrough as evidenced by the approval of the BNT162b2 vaccine for prevention of coronavirus disease 2019 (COVID-19) in 2021. Compared with conventional cationic gene vectors, IDDSs can decrease the toxicity of carriers to cell membranes, and increase cellular uptake and endosomal escape of nucleic acids by their unique pH-responsive structures. Despite the progress, there remain necessary requirements for designing more efficient IDDSs for precise gene therapy. Herein, we systematically classify the IDDSs and summarize the characteristics and advantages of IDDSs in order to explore the underlying design mechanisms. The delivery mechanisms and therapeutic applications of IDDSs are comprehensively reviewed for the delivery of pDNA and four kinds of RNA. In particular, organ selecting considerations and high-throughput screening are highlighted to explore efficiently multifunctional ionizable nanomaterials with superior gene delivery capacity. We anticipate providing references for researchers to rationally design more efficient and accurate targeted gene delivery systems in the future, and indicate ideas for developing next generation gene vectors.
Collapse
Affiliation(s)
- Yu-Qi Zhang
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Ran-Ran Guo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Yong-Hu Chen
- School of Pharmacy, Yanbian University, Yanji, 133002, Jilin, China
| | - Tian-Cheng Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Wen-Zhen Du
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Rong-Wu Xiang
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Ji-Bin Guan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yu-Peng Li
- Masonic Cancer Center and Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yuan-Yu Huang
- Advanced Research Institute of Multidisciplinary Science; School of Life Science; School of Medical Technology; Key Laboratory of Molecular Medicine and Biotherapy; Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhi-Qiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, 523018, Guangdong, China
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China.
| | - Gui-Xia Ling
- Faculty of Medical Device, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
17
|
Hari SK, Gauba A, Shrivastava N, Tripathi RM, Jain SK, Pandey AK. Polymeric micelles and cancer therapy: an ingenious multimodal tumor-targeted drug delivery system. Drug Deliv Transl Res 2023; 13:135-163. [PMID: 35727533 DOI: 10.1007/s13346-022-01197-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/13/2022]
Abstract
Since the beginning of pharmaceutical research, drug delivery methods have been an integral part of it. Polymeric micelles (PMs) have emerged as multifunctional nanoparticles in the current technological era of nanocarriers, and they have shown promise in a range of scientific fields. They can alter the release profile of integrated pharmacological substances and concentrate them in the target zone due to their improved permeability and retention, making them more suitable for poorly soluble medicines. With their ability to deliver poorly soluble chemotherapeutic drugs, PMs have garnered considerable interest in cancer. As a result of their remarkable biocompatibility, improved permeability, and minimal toxicity to healthy cells, while also their capacity to solubilize a wide range of drugs in their micellar core, PMs are expected to be a successful treatment option for cancer therapy in the future. Their nano-size enables them to accumulate in the tumor microenvironment (TME) via the enhanced permeability and retention (EPR) effect. In this review, our major aim is to focus primarily on the stellar applications of PMs in the field of cancer therapeutics along with its mechanism of action and its latest advancements in drug and gene delivery (DNA/siRNA) for cancer, using various therapeutic strategies such as crossing blood-brain barrier, gene therapy, photothermal therapy (PTT), and immunotherapy. Furthermore, PMs can be employed as "smart drug carriers," allowing them to target specific cancer sites using a variety of stimuli (endogenous and exogenous), which improve the specificity and efficacy of micelle-based targeted drug delivery. All the many types of stimulants, as well as how the complex of PM and various anticancer drugs react to it, and their pharmacodynamics are also reviewed here. In conclusion, commercializing engineered micelle nanoparticles (MNPs) for application in therapy and imaging can be considered as a potential approach to improve the therapeutic index of anticancer drugs. Furthermore, PM has stimulated intense interest in research and clinical practice, and in light of this, we have also highlighted a few PMs that have previously been approved for therapeutic use, while the majority are still being studied in clinical trials for various cancer therapies.
Collapse
Affiliation(s)
- Sharath Kumar Hari
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India
| | - Ankita Gauba
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India
| | - Neeraj Shrivastava
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India
| | - Ravi Mani Tripathi
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India.
| | - Sudhir Kumar Jain
- School of Studies in Microbiology, Vikram University, Ujjain, Madhya Pradesh, 456010, India
| | - Akhilesh Kumar Pandey
- Department of Biological Sciences, Rani Durgavati University, Jabalpur, M.P, 482001, India.,Vikram University, Ujjain, Madhya Pradesh, 456010, India
| |
Collapse
|
18
|
Wang X, Yang X, Hu Y, Guo S, Lu J. Bionic Assembly of Layered Double Hydroxides Nanosheets and Positively Charged Micelles by Counterions Balance and Their Selective Detection of Mannose. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:14686-14694. [PMID: 36437733 DOI: 10.1021/acs.langmuir.2c02225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The selective detection of mannose is significant for tumor early diagnosis. However, current methods for detecting mannose are expensive and time-consuming, limiting their application. In this paper, we have obtained a 25-layer positively charged micellar/LDHs nanocomposite film system by electrostatic layer-by-layer assembly with reference to the unique properties of homogeneous charge ion attraction and charge overcompensation in biomolecules: hexadecyl trimethylammonium bromide (CTAB) was used to coat neutral molecules of fluorescein (FLU) to form (FLU@CTAB) cationic micelles, which were electrostatically assembled with laminate positively charged layered double hydroxides (LDHs) nanosheets to form (FLU@CTAB/LDHs)n ultrathin films (UTFs) by the layer-by-layer electrostatic assembly, where the mediating role of the Br- counteranion had a profound effect on the success of the assembly. Moreover, compared to pure FLU solution, the fluorescence intensity and the lifetime of (FLU@CTAB/LDHs)20 UTFs were enhanced by 1.6 and 2 times, respectively. (FLU@CTAB/LDHs)20 UTFs exhibited selective detection for d-mannose with a detection limit of 0.05 mg·mL-1. Therefore, the (FLU@CTAB/LDHs)n UTFs can be a novel biosensor. Compared to conventional powder sensors, (FLU@CTAB/LDHs)n thin-film fluorescent sensors are more promising for device implementation. Moreover, the design strategy of positively charged micellar/LDHs nanocomposite systems breaks the current limitation that LDHs can only be assembled with anions or neutral molecules and extends the scope of counterion-mediated host-guest to the nanosheet-micellar system.
Collapse
Affiliation(s)
- Xiaoyan Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, P.O. Box 98, Beisanhuan East Road 15, Beijing, 100029, China
| | - Xueting Yang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, P.O. Box 98, Beisanhuan East Road 15, Beijing, 100029, China
| | - Yuehua Hu
- Shanghai Research Institute Sinopec Lubricant, 455 Gaoyang Road, Shanghai, 200080, China
| | - Shuaitian Guo
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, P.O. Box 98, Beisanhuan East Road 15, Beijing, 100029, China
| | - Jun Lu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, P.O. Box 98, Beisanhuan East Road 15, Beijing, 100029, China
- Beijing Advanced Innovation Center for Soft Mater Science and Engineering, P.O. Box 98, Beisanhuan East Road 15, Beijing, 100029, China
| |
Collapse
|
19
|
Hanson MG, Grimme CJ, Santa Chalarca CF, Reineke TM. Cationic Micelles Outperform Linear Polymers for Delivery of Antisense Oligonucleotides in Serum: An Exploration of Polymer Architecture, Cationic Moieties, and Cell Addition Order. Bioconjug Chem 2022; 33:2121-2131. [PMID: 36265078 DOI: 10.1021/acs.bioconjchem.2c00379] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Antisense oligonucleotides (ASOs) are an important emerging therapeutic; however, they struggle to enter cells without a delivery vehicle, such as a cationic polymer. To understand the role of polymer architecture for ASO delivery, five linear polymers and five diblock polymers (capable of self-assembly into micelles) were synthesized with varying cationic groups. After complexation of each polymer/micelle with ASO, it was found that less bulky cationic moieties transfected the ASO more effectively. Interestingly, however the ASO internalization trend was the opposite of the transfection trend for cationic moiety, indicating internalization is not the major factor in determining transfection efficiency for this series. Micelleplexes (micelle-ASO complexes) generally enable higher transfection efficacy as compared to polyplexes (linear polymer-ASO complexes). Additionally, the order of addition of cells and complexes was explored. Linear polyplexes showed better transfection efficiency in adhered cells, whereas micelleplexes delivered the ASO more efficiently when the cells and micelleplexes were added simultaneously. This phenomenon may be due to increased cell-complex interactions as micelleplexes have increased colloidal stability compared to polyplexes. These findings emphasize the importance of polymer composition and architecture in governing the cellular interactions necessary for transfection, thus allowing advancement in the design principles for nonviral nucleic acid delivery formulations.
Collapse
Affiliation(s)
- Mckenna G Hanson
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421Washington Avenue SE, Minneapolis, Minnesota 55455, United States
| | - Cristiam F Santa Chalarca
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
20
|
Cyclodextrin-based dermatological formulations: dermopharmaceutical and cosmetic applications. Colloids Surf B Biointerfaces 2022; 221:113012. [DOI: 10.1016/j.colsurfb.2022.113012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
|
21
|
Huang S, Hao XY, Li YJ, Wu JY, Xiang DX, Luo S. Nonviral delivery systems for antisense oligonucleotide therapeutics. Biomater Res 2022; 26:49. [PMID: 36180936 PMCID: PMC9523189 DOI: 10.1186/s40824-022-00292-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
Antisense oligonucleotides (ASOs) are an important tool for the treatment of many genetic disorders. However, similar to other gene drugs, vectors are often required to protect them from degradation and clearance, and to accomplish their transport in vivo. Compared with viral vectors, artificial nonviral nanoparticles have a variety of design, synthesis, and formulation possibilities that can be selected to accomplish protection and delivery for specific applications, and they have served critical therapeutic purposes in animal model research and clinical applications, allowing safe and efficient gene delivery processes into the target cells. We believe that as new ASO drugs develop, the exploration for corresponding nonviral vectors is inevitable. Intensive development of nonviral vectors with improved delivery strategies based on specific targets can continue to expand the value of ASO therapeutic approaches. Here, we provide an overview of current nonviral delivery strategies, including ASOs modifications, action mechanisms, and multi-carrier methods, which aim to address the irreplaceable role of nonviral vectors in the progressive development of ASOs delivery.
Collapse
Affiliation(s)
- Si Huang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, People's Republic of China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xin-Yan Hao
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, People's Republic of China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yong-Jiang Li
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, People's Republic of China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Jun-Yong Wu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, People's Republic of China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Da-Xiong Xiang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China.,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, People's Republic of China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Shilin Luo
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China. .,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, People's Republic of China. .,Institute of Clinical Pharmacy, Central South University, Changsha, China.
| |
Collapse
|
22
|
Pasha SS, Banerjee A, Sreedharan S, Singh S, Kandoth N, Vallis KA, Pal SK, Pramanik SK, Das A. Ultrasensitive Reagent for Ratiometric Detection and Detoxification of iAsIII in Water and Mitochondria. Inorg Chem 2022; 61:13115-13124. [PMID: 35950896 DOI: 10.1021/acs.inorgchem.2c01761] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Toxicity induced by inorganic arsenic as AsO33- (iAsIII) is of global concern. Reliable detection of the maximum allowed contaminant level for arsenic in drinking water and in the cellular system remains a challenge for the water quality management and assessment of toxicity in the cellular milieu, respectively. A new Ir(III)-based phosphorescent molecule (AS-1; λExt = 415 nm and λEms = 600 nm, Φ = 0.3) is synthesized for the selective detection of iAsIII in an aqueous solution with a ratiometric luminescence response even in the presence of iAsV and all other common inorganic cations and anions. The relatively higher affinity of the thioimidazole ligand (HPBT) toward iAsIII led to the formation of a fluorescent molecule iAsV-HPBT (λExt = 415 nm and λEms = 466 nm, Φ = 0.28) for the reaction of iAsIII and AS-1. An improved limit of quantitation (LOQ) down to 0.2 ppb is achieved when AS-1 is used in the CTAB micellar system. Presumably, the cationic surfactants favor the localization of AS-1@CTABMicelle in mitochondria of MCF7 cells, and this is confirmed from the images of the confocal laser fluorescence scanning microscopic studies. Importantly, cell viability assay studies confirm that AS-1@CTABMicelle induces dose-dependent detoxification of iAsIII in live cells. Further, luminescence responses at 466 nm could be utilized for developing a hand-held device for the in-field application. Such a reagent that allows for ratiometric detection of iAsIII with LOQ of 2.6 nM (0.5 ppb) in water, as well as helps in visualizing its distribution in mitochondria with a detoxifying effect, is rather unique in contemporary literature.
Collapse
Affiliation(s)
- Sheik Saleem Pasha
- Central Salt and Marine Chemicals Research Institute, Bhavnagar 364002, Gujarat, India
| | - Amrita Banerjee
- Technical Research Centre, Department of Chemical, Biological & Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, West Bengal 700106, India
| | - Sreejesh Sreedharan
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, U.K.,Human Sciences Research Centre, University of Derby, Derby DE221GB, U.K
| | - Soumendra Singh
- Technical Research Centre, Department of Chemical, Biological & Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, West Bengal 700106, India
| | - Noufal Kandoth
- Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India
| | - Katherine A Vallis
- Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, U.K
| | - Samir Kumar Pal
- Technical Research Centre, Department of Chemical, Biological & Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, West Bengal 700106, India
| | - Sumit Kumar Pramanik
- Central Salt and Marine Chemicals Research Institute, Bhavnagar 364002, Gujarat, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amitava Das
- Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, West Bengal, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
23
|
Soh WWM, Teoh RYP, Zhu J, Xun Y, Wee CY, Ding J, Thian ES, Li J. Facile Construction of a Two-in-One Injectable Micelleplex-Loaded Thermogel System for the Prolonged Delivery of Plasmid DNA. Biomacromolecules 2022; 23:3477-3492. [PMID: 35878156 DOI: 10.1021/acs.biomac.2c00648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Nanoparticle-hydrogel systems have recently emerged as a class of interesting hybrid materials with immense potential for several biomedical applications. Remarkably, the incorporation of nanoparticles into a hydrogel may yield synergistic benefits lacking in a singular system. However, most synthetic strategies require laborious steps to achieve the system, severely restricting the process of translational research. Herein, a facile strategy to access a two-in-one system comprising two distinct polyurethane (PU)-based micellar systems is demonstrated and applied as a novel sustained gene delivery platform, where the two PUs are synthesized similarly but with slightly different compositions. One PU forms cationic micelles that complex with plasmid DNA (pDNA), which are loaded into a thermogel formed by another PU micellar system for the prolonged release of pDNA micelleplexes. Specifically, a thermogelling multiblock PU copolymer (denoted as EPH) was synthesized via the step-growth polymerization of poly(ethylene glycol), poly(propylene glycol), and poly(3-hydroxybutyrate). By further introducing a cationic extender, 3-(dimethylamino)-1,2-propanediol, into the reaction feed, a series of cationic PUs (denoted as EPHD) with varying compositions were obtained. The EPHDs formed positively charged micelles in aqueous solutions, efficiently condensed pDNA into nano-sized micelleplexes (<200 nm) at optimized w/w ratios, and mediated transient green fluorescence protein expression in HEK293T cells at 48 h post-transfection. On the other hand, aqueous EPH solution (4 wt %) was injectable at 4 °C and rapidly gelled upon heating to 37 °C to form a stable hydrogel depot. EPHD/pDNA micelleplexes were easily loaded into EPH by mixing the solutions at 4 °C, before heating to 37 °C, leading to the resultant hydrogel system. The in vitro release study revealed that while free pDNA loaded in the thermogel was completely released in 2 weeks, the release of EPHD/pDNA micelleplexes was prolonged to at least 28 days, suggesting substantial micelleplex-hydrogel interactions. Intact, bioactive, and noncytotoxic EPHD/pDNA micelleplexes in the release media were proved by gel retardation, in vitro gene transfection, and CCK-8 cytotoxicity assay results, respectively. Collectively, this work presents a simple approach to achieving and optimizing a novel two-in-one nanoparticle-hydrogel system for the prolonged delivery of pDNA and may be promising for long-term gene delivery applications.
Collapse
Affiliation(s)
- Wilson Wee Mia Soh
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Rachel Yun Pei Teoh
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jingling Zhu
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore.,NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Yanran Xun
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Chien Yi Wee
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Jun Ding
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Eng San Thian
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Jun Li
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore.,NUS Environmental Research Institute (NERI), National University of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| |
Collapse
|
24
|
Grimme CJ, Hanson MG, Corcoran LG, Reineke TM. Polycation Architecture Affects Complexation and Delivery of Short Antisense Oligonucleotides: Micelleplexes Outperform Polyplexes. Biomacromolecules 2022; 23:3257-3271. [PMID: 35862267 DOI: 10.1021/acs.biomac.2c00338] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Herein, we examine the complexation and biological delivery of a short single-stranded antisense oligonucleotide (ASO) payload with four polymer derivatives that form two architectural variants (polyplexes and micelleplexes): a homopolymer poly(2-dimethylaminoethyl methacrylate) (D), a diblock polymer poly(ethylene glycol)methylether methacrylate-block-poly(2-dimethylaminoethyl methacrylate) (ObD), and two micelle-forming variants, poly(2-dimethylaminoethyl methacrylate)-block-poly(n-butyl methacrylate) (DB) and poly(ethylene glycol)methylether methacrylate-block-poly(2-dimethylaminoethyl methacrylate)-block-poly(n-butyl methacrylate) (ObDB). Both polyplexes and micelleplexes complexed ASOs, and the incorporation of an Ob brush enhances colloidal stability. Micellplexes are templated by the size and shape of the unloaded micelle and that micelle-ASO complexation is not sensitive to formulation/mixing order, allowing ease, versatility, and reproducibility in packaging short oligonucleotides. The DB micelleplexes promoted the largest gene silencing, internalization, and tolerable toxicity while the ObDB micelleplexes displayed enhanced colloidal stability and highly efficient payload trafficking despite having lower cellular uptake. Overall, this work demonstrates that cationic micelles are superior delivery vehicles for ASOs denoting the importance of vehicle architecture in biological performance.
Collapse
Affiliation(s)
- Christian J Grimme
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Avenue SE, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Louis G Corcoran
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
25
|
Akib AA, Shakil R, Rumon MMH, Roy CK, Chowdhury EH, Chowdhury AN. Natural and Synthetic Micelles for Delivery of Small Molecule Drugs, Imaging Agents and Nucleic Acids. Curr Pharm Des 2022; 28:1389-1405. [PMID: 35524674 DOI: 10.2174/1381612828666220506135301] [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: 10/05/2021] [Accepted: 02/02/2022] [Indexed: 11/22/2022]
Abstract
The poor solubility, lack of targetability, quick renal clearance, and degradability of many therapeutic and imaging agents strongly limit their applications inside the human body. Amphiphilic copolymers having self-assembling properties can form core-shell structures called micelles, a promising nanocarrier for hydrophobic drugs, plasmid DNA, oligonucleotides, small interfering RNAs (siRNAs) and imaging agents. Fabrication of micelles loaded with different pharmaceutical agents provides numerous advantages including therapeutic efficacy, diagnostic sensitivity, and controlled release to the desired tissues. Moreover, due to their smaller particle size (10-100 nm) and modified surfaces with different functional groups (such as ligands) help them to accumulate easily in the target location, enhancing cellular uptake and reducing unwanted side effects. Furthermore, the release of the encapsulated agents may also be triggered from stimuli-sensitive micelles at different physiological conditions or by an external stimulus. In this review article, we discuss the recent advancement in formulating and targeting different natural and synthetic micelles including block copolymer micelles, cationic micelles, and dendrimers-, polysaccharide- and protein-based micelles for the delivery of different therapeutic and diagnostic agents. Finally, their applications, outcomes, and future perspectives have been summarized.
Collapse
Affiliation(s)
- Anwarul Azim Akib
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Ragib Shakil
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Md Mahamudul Hasan Rumon
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Chanchal Kumar Roy
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Malaysia
| | - Al-Nakib Chowdhury
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| |
Collapse
|
26
|
Faria R, Paul M, Biswas S, Vivès E, Boisguérin P, Sousa Â, Costa D. Peptides vs. Polymers: Searching for the Most Efficient Delivery System for Mitochondrial Gene Therapy. Pharmaceutics 2022; 14:757. [PMID: 35456591 PMCID: PMC9026848 DOI: 10.3390/pharmaceutics14040757] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Together with the nucleus, the mitochondrion has its own genome. Mutations in mitochondrial DNA are responsible for a variety of disorders, including neurodegenerative diseases and cancer. Current therapeutic approaches are not effective. In this sense, mitochondrial gene therapy emerges as a valuable and promising therapeutic tool. To accomplish this goal, the design/development of a mitochondrial-specific gene delivery system is imperative. In this work, we explored the ability of novel polymer- and peptide-based systems for mitochondrial targeting, gene delivery, and protein expression, performing a comparison between them to reveal the most adequate system for mitochondrial gene therapy. Therefore, we synthesized a novel mitochondria-targeting polymer (polyethylenimine-dequalinium) to load and complex a mitochondrial-gene-based plasmid. The polymeric complexes exhibited physicochemical properties and cytotoxic profiles dependent on the nitrogen-to-phosphate-group ratio (N/P). A fluorescence confocal microscopy study revealed the mitochondrial targeting specificity of polymeric complexes. Moreover, transfection mediated by polymer and peptide delivery systems led to gene expression in mitochondria. Additionally, the mitochondrial protein was produced. A comparative study between polymeric and peptide/plasmid DNA complexes showed the great capacity of peptides to complex pDNA at lower N/P ratios, forming smaller particles bearing a positive charge, with repercussions on their capacity for cellular transfection, mitochondria targeting and, ultimately, gene delivery and protein expression. This report is a significant contribution to the implementation of mitochondrial gene therapy, instigating further research on the development of peptide-based delivery systems towards clinical translation.
Collapse
Affiliation(s)
- Rúben Faria
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilha, Portugal; (R.F.); (Â.S.)
| | - Milan Paul
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, India; (M.P.); (S.B.)
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, India; (M.P.); (S.B.)
| | - Eric Vivès
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34295 Montpellier, France; (E.V.); (P.B.)
| | - Prisca Boisguérin
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34295 Montpellier, France; (E.V.); (P.B.)
| | - Ângela Sousa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilha, Portugal; (R.F.); (Â.S.)
| | - Diana Costa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilha, Portugal; (R.F.); (Â.S.)
| |
Collapse
|
27
|
Zhai H, Chen K, Meng Y, Wu Z, Deng R, Bai Y, Zhou J, Quan D. Synthesis and self-assembly of amphiphilic diblock polycarbonates with various pendant hydrophilic groups. POLYMER 2022. [DOI: 10.1016/j.polymer.2022.124664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
28
|
Zhang P, Chen D, Li L, Sun K. Charge reversal nano-systems for tumor therapy. J Nanobiotechnology 2022; 20:31. [PMID: 35012546 PMCID: PMC8751315 DOI: 10.1186/s12951-021-01221-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022] Open
Abstract
Surface charge of biological and medical nanocarriers has been demonstrated to play an important role in cellular uptake. Owing to the unique physicochemical properties, charge-reversal delivery strategy has rapidly developed as a promising approach for drug delivery application, especially for cancer treatment. Charge-reversal nanocarriers are neutral/negatively charged at physiological conditions while could be triggered to positively charged by specific stimuli (i.e., pH, redox, ROS, enzyme, light or temperature) to achieve the prolonged blood circulation and enhanced tumor cellular uptake, thus to potentiate the antitumor effects of delivered therapeutic agents. In this review, we comprehensively summarized the recent advances of charge-reversal nanocarriers, including: (i) the effect of surface charge on cellular uptake; (ii) charge-conversion mechanisms responding to several specific stimuli; (iii) relation between the chemical structure and charge reversal activity; and (iv) polymeric materials that are commonly applied in the charge-reversal delivery systems.
Collapse
Affiliation(s)
- Peng Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong, People's Republic of China.
| | - Daoyuan Chen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong, People's Republic of China
| | - Lin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong, People's Republic of China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, 30 Qingquan Road, Yantai, 264005, Shandong, People's Republic of China.,State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co. Ltd, Yantai, 264003, People's Republic of China
| |
Collapse
|
29
|
Iqbal S, Zhao Z. Poly (β amino esters) copolymers: Novel potential vectors for delivery of genes and related therapeutics. Int J Pharm 2022; 611:121289. [PMID: 34775041 DOI: 10.1016/j.ijpharm.2021.121289] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/26/2021] [Accepted: 11/08/2021] [Indexed: 12/21/2022]
Abstract
The unique properties of polymers have performed an essential contribution to the drug delivery system by providing an outstanding platform for the delivery of macromolecules and genes. However, the block copolymers have been the subject of many recently published works whose results have demonstrated excellent performance in drug targeting. Poly(β-amino esters) (PβAEs) copolymers are the synthetic cationic polymers that are tailored by chemically joining PβAEs with other additives to demonstrate extraordinary efficiency in designing pre-defined and pre-programmed nanostructures, site-specific delivery, andovercoming the distinct cellular barriers. Different compositional and structural libraries could be generated by combinatorial chemistry and by the addition of various novel functional additives that fulfill the multiple requirements of targeted delivery. These intriguing attributes allow PβAE-copolymers to have customized therapeutic functions such as excellent encapsulation capacity, high stability, and stimuli-responsive release. Here, we give an overview of PβAE copolymers-based formulations along with focusing on most notable improvements such as structural modifications, bio-conjugations, and stimuli-responsive approaches, for safe and effective nucleic acids delivery.
Collapse
Affiliation(s)
- Sajid Iqbal
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China
| | - Zhongxi Zhao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China; Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan, Shandong 250012, PR China; Pediatric Pharmaceutical Engineering Laboratory of Shandong Province, Shandong Dyne Marine Biopharmaceutical Company Limited, Rongcheng, Shandong 264300, PR China; Chemical Immunopharmaceutical Engineering Laboratory of Shandong Province, Shandong Xili Pharmaceutical Company Limited, Heze, Shandong 274300, PR China.
| |
Collapse
|
30
|
Ji W, Li Y, Peng H, Zhao R, Shen J, Wu Y, Wang J, Hao Q, Lu Z, Yang J, Zhang X. Self-Catalytic Small Interfering RNA Nanocarriers for Synergistic Treatment of Neurodegenerative Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105711. [PMID: 34601753 DOI: 10.1002/adma.202105711] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/14/2021] [Indexed: 06/13/2023]
Abstract
Gene therapy has shown great potential for neurodegenerative diseases with complex pathology. However, its therapeutic effect is limited due to the delivery barriers and its own single function. Herein, self-catalytic small interfering RNA (siRNA) nanocarriers (S/Ce-PABMS) are developed to catalyze delivery process and treatment process for synergistic treatment of neurodegenerative diseases. On the one hand, the rough surface of the S/Ce-PABMS mediated by ceria (CeO2 ) nanozymes can catalyze cellular uptake in the delivery process, so that S/Ce-PABMS with acetylcholine analogs penetrate the blood-brain barrier and enter neurons more effectively. On the other hand, the CeO2 nanozymes can catalyze the treatment process by scavenging excess reactive oxygen species, and cooperate with siRNA-targeting SNCA to decrease the α-synuclein (α-syn) aggregation and alleviate the Parkinsonian pathology. Moreover, the S/Ce-PABMS treatment reduces the number of activated microglia and regulates the release of inflammatory cytokine, thereby relieving neuroinflammation. After treatment with S/Ce-PABMS, dyskinesia in Parkinson's disease model mice is significantly alleviated. The finding shows that the self-catalytic nanocarriers, S/Ce-PABMS, have great potential in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Weihong Ji
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yan Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, P. R. China
| | - Huan Peng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ruichen Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jie Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yanyue Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jianze Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Qiulian Hao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Zhiguo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Jun Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
31
|
Dual-modified nanoparticles overcome sequential absorption barriers for oral insulin delivery. J Control Release 2021; 342:1-13. [PMID: 34864116 DOI: 10.1016/j.jconrel.2021.11.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 01/25/2023]
Abstract
The efficacy of oral insulin drug delivery is seriously hampered by multiple gastrointestinal barriers, especially transepithelial barriers, including apical endocytosis, lysosomal degradation, cytosolic diffusion and basolateral exocytosis. In this study, a functional nanoparticle (PG-FAPEP) with dual-modification was constructed to sequentially address these important absorption obstacles for improved oral insulin delivery. The dual surface decorations folate and charge-convertible tripeptide endowed PG-FAPEP with the ability to target the apical and basolateral sides of enterocytes, respectively. After fast diffusion across the mucus layer, PG-FAPEP could be efficiently internalized into epithelial cells via a folate receptor-mediated pathway and subsequently became positively charged in acidic lysosomes due to the surface tripeptide, triggering the proton sponge effect to escape lysosomes. When entering the cytosolic medium, PG-FAPEP was converted to neutral charge again, attenuating intracellular adhesion, and gained improved motility toward the basolateral side. Finally, the tripeptide helped PG-FAPEP recognize the proton-coupled oligopeptide transporter (PHT1) in the basolateral membrane, boosting intact exocytosis across intestinal epithelial cells. The in vivo studies further verified that PG-FAPEP could traverse the intestinal epithelium by folate receptor-mediated endocytosis, lysosomal escape, and PHT1-mediated exocytosis, exhibiting a high oral insulin bioavailability of 14.3% and a prolonged hypoglycemic effect. This formulation addresses multiple absorption barriers on demand with a simple dual-modification strategy. Therefore, these features allow PG-FAPEP to unleash the potential of oral macromolecule delivery.
Collapse
|
32
|
Zheng B, Zhang P, Wang H, Wang J, Liu ZH, Zhang D. Advances in Research on Bladder Cancer Targeting Peptides: a Review. Cell Biochem Biophys 2021; 79:711-718. [PMID: 34468956 PMCID: PMC8558283 DOI: 10.1007/s12013-021-01019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2021] [Indexed: 12/04/2022]
Abstract
Bladder cancer (Bca) is the second most common malignant tumor of the genitourinary system in Chinese male population with high potential of recurrence and progression. The overall prognosis has not been improved significantly for the past 30 years due to the lack of early theranostic technique. Currently the early theranostic technique for bladder cancer is mainly through the intravesical approach, but the clinical outcomes are poor due to the limited tumor-targeting efficiency. Therefore, the targeting peptides for bladder cancer provide possibility to advance intravesical theranostic technique. However, no systematic review has covered the wide use of the targeting peptides for intravesical theranostic techniques in bladder cancer. Herein, a summary of original researches introduces all aspects of the targeting peptides for bladder cancer, including the peptide screening, the targeting mechanism and its preclinical application.
Collapse
Affiliation(s)
- Bin Zheng
- Zhejiang Chinese Medical University, 310053, HangZhou, China
- Zhejiang Provincial People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
| | - Pu Zhang
- Zhejiang Provincial People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
| | - Heng Wang
- Zhejiang Provincial People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
| | - Jinxue Wang
- Handan Central hospital, 056001, Handan, China
| | - Zheng Hong Liu
- Zhejiang Chinese Medical University, 310053, HangZhou, China
| | - DaHong Zhang
- Zhejiang Chinese Medical University, 310053, HangZhou, China.
| |
Collapse
|
33
|
Er S, Laraib U, Arshad R, Sargazi S, Rahdar A, Pandey S, Thakur VK, Díez-Pascual AM. Amino Acids, Peptides, and Proteins: Implications for Nanotechnological Applications in Biosensing and Drug/Gene Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:3002. [PMID: 34835766 PMCID: PMC8622868 DOI: 10.3390/nano11113002] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022]
Abstract
Over various scientific fields in biochemistry, amino acids have been highlighted in research works. Protein, peptide- and amino acid-based drug delivery systems have proficiently transformed nanotechnology via immense flexibility in their features for attaching various drug molecules and biodegradable polymers. In this regard, novel nanostructures including carbon nanotubes, electrospun carbon nanofibers, gold nanoislands, and metal-based nanoparticles have been introduced as nanosensors for accurate detection of these organic compounds. These nanostructures can bind the biological receptor to the sensor surface and increase the surface area of the working electrode, significantly enhancing the biosensor performance. Interestingly, protein-based nanocarriers have also emerged as useful drug and gene delivery platforms. This is important since, despite recent advancements, there are still biological barriers and other obstacles limiting gene and drug delivery efficacy. Currently available strategies for gene therapy are not cost-effective, and they do not deliver the genetic cargo effectively to target sites. With rapid advancements in nanotechnology, novel gene delivery systems are introduced as nonviral vectors such as protein, peptide, and amino acid-based nanostructures. These nano-based delivery platforms can be tailored into functional transformation using proteins and peptides ligands based nanocarriers, usually overexpressed in the specified diseases. The purpose of this review is to shed light on traditional and nanotechnology-based methods to detect amino acids, peptides, and proteins. Furthermore, new insights into the potential of amino protein-based nanoassemblies for targeted drug delivery or gene transfer are presented.
Collapse
Affiliation(s)
- Simge Er
- Biochemistry Department, Faculty of Science, Ege University, Bornova-Izmir 35100, Turkey;
| | - Ushna Laraib
- Department of Pharmacy, College of Pharmacy, University of Sargodha, Sargodha 40100, Pakistan;
| | - Rabia Arshad
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran;
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 538-98615, Iran
| | - Sadanand Pandey
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea;
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Centre, Scotland’s Rural College (SRUC), Kings Buildings, Edinburgh EH9 3JG, UK;
- School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, Uttarakhand, India
| | - Ana M. Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain
| |
Collapse
|
34
|
Targeted delivery of miR-218 via decorated hyperbranched polyamidoamine for liver cancer regression. Int J Pharm 2021; 610:121256. [PMID: 34732362 DOI: 10.1016/j.ijpharm.2021.121256] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/15/2021] [Accepted: 10/28/2021] [Indexed: 01/28/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of most common causes of cancer death worldwide. MicroRNA (miRNA) replacement gene therapy is a novel approach for HCC management. MiR-218 is a promising tumor suppressor miRNA that is down-regulated in HCC. Here, our aim was the targeted delivery of miR-218 expressing DNA plasmid (pmiR-218) to suppress HCC in vitro and in vivo. Hyperbranched polyamidoamine was synthesized via simple and economically one-pot reaction followed by decoration with lactobionic acid (LA-PAMAM) to selectively deliver and restore miR-218 expression in HCC. In vitro cytotoxicity investigations revealed the high biocompatibility of LA-PAMAM. Furthermore, decoration of hyperbranched polymer with LA moieties enabled LA-PAMAM to deliver pmiR-218 more efficiently to HepG2 cells compared to both PMAMA and naked pmiR-218. Such efficient delivery of miR-218 resulted in suppression of HepG2 proliferation and down-regulation of its oncogenic HOXA1 target. In vivo, LA-PAMAM/pmiR-218 treatment of HCC induced by DEN and CCl4 in mice leads to an obvious decrease in the number and size of HCC nodules. In addition, LA-PAMAM/pmiR-218 significantly improved the liver histological features, as well as down-regulated the HOXA1 in liver tissue. In conclusion, this study showed the potential of LA-PAMAM carrier for the targeted delivery of tumor suppressor miR-218 as a therapeutic candidate for HCC.
Collapse
|
35
|
Richter F, Leer K, Martin L, Mapfumo P, Solomun JI, Kuchenbrod MT, Hoeppener S, Brendel JC, Traeger A. The impact of anionic polymers on gene delivery: how composition and assembly help evading the toxicity-efficiency dilemma. J Nanobiotechnology 2021; 19:292. [PMID: 34579715 PMCID: PMC8477462 DOI: 10.1186/s12951-021-00994-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/10/2021] [Indexed: 02/02/2023] Open
Abstract
Cationic polymers have been widely studied for non-viral gene delivery due to their ability to bind genetic material and to interact with cellular membranes. However, their charged nature carries the risk of increased cytotoxicity and interaction with serum proteins, limiting their potential in vivo application. Therefore, hydrophilic or anionic shielding polymers are applied to counteract these effects. Herein, a series of micelle-forming and micelle-shielding polymers were synthesized via RAFT polymerization. The copolymer poly[(n-butyl acrylate)-b-(2-(dimethyl amino)ethyl acrylamide)] (P(nBA-b-DMAEAm)) was assembled into cationic micelles and different shielding polymers were applied, i.e., poly(acrylic acid) (PAA), poly(4-acryloyl morpholine) (PNAM) or P(NAM-b-AA) block copolymer. These systems were compared to a triblock terpolymer micelle comprising PAA as the middle block. The assemblies were investigated regarding their morphology, interaction with pDNA, cytotoxicity, transfection efficiency, polyplex uptake and endosomal escape. The naked cationic micelle exhibited superior transfection efficiency, but increased cytotoxicity. The addition of shielding polymers led to reduced toxicity. In particular, the triblock terpolymer micelle convinced with high cell viability and no significant loss in efficiency. The highest shielding effect was achieved by layering micelles with P(NAM-b-AA) supporting the colloidal stability at neutral zeta potential and completely restoring cell viability while maintaining moderate transfection efficiencies. The high potential of this micelle-layer-combination for gene delivery was illustrated for the first time.
Collapse
Affiliation(s)
- Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Katharina Leer
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Liam Martin
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Prosper Mapfumo
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jana I Solomun
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Maren T Kuchenbrod
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany.
| |
Collapse
|
36
|
Development of Tailor-Made Dendrimer Ternary Complexes for Drug/Gene Co-Delivery in Cancer. Pharmaceutics 2021; 13:pharmaceutics13081256. [PMID: 34452218 PMCID: PMC8401607 DOI: 10.3390/pharmaceutics13081256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 12/03/2022] Open
Abstract
Cancer gene therapy, mediated by non-viral systems, remains a major research focus. To contribute to this field, in this work we reported on the development of dendrimer drug/gene ternary complexes. This innovative approach explored the great capacity of both polyamidoamine (PAMAM)-paclitaxel (PTX) conjugate and polyethylenimine (PEI) polymers to complex a p53-encoding plasmid DNA (pDNA), highlighting the utility of considering two compacting agents. The pDNA complexation capacity has been investigated as function of the nitrogen to phosphate groups ratio (N/P), which revealed to be a tailoring parameter. The physicochemical properties of the conceived ternary complexes were revealed and were found to be promising for cellular transfection. Furthermore, the formulated co-delivery systems demonstrated to be biocompatible. The ternary systems were able of cellular internalization and payload intracellular release. Confocal microscopy studies showed the co-localization of stained pDNA with the nucleus of cancer cells, after transfection mediated by these carriers. From this achievement, p53 gene expression occurred with the production of protein. Moreover, the activation of caspase-3 indicated apoptosis of cancer cells. This work represents a great progress on the design of dendrimer drug/gene co-delivery systems towards a more efficient cancer therapy. In this way, it instigates further in vitro studies concerning the evaluation of their therapeutic potential, expectedly supported by the synergistic effect, in tumoral cells.
Collapse
|
37
|
Faria R, Vivés E, Boisguerin P, Sousa A, Costa D. Development of Peptide-Based Nanoparticles for Mitochondrial Plasmid DNA Delivery. Polymers (Basel) 2021; 13:1836. [PMID: 34206125 PMCID: PMC8199553 DOI: 10.3390/polym13111836] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 12/25/2022] Open
Abstract
A mitochondrion is a cellular organelle able to produce cellular energy in the form of adenosine triphosphate (ATP). As in the nucleus, mitochondria contain their own genome: the mitochondrial DNA (mtDNA). This genome is particularly susceptible to mutations that are at the basis of a multitude of disorders, especially those affecting the heart, the central nervous system and muscles. Conventional clinical practice applied to mitochondrial diseases is very limited and ineffective; a clear need for innovative therapies is demonstrated. Gene therapy seems to be a promising approach. The use of mitochondrial DNA as a therapeutic, optimized by peptide-based complexes with mitochondrial targeting, can be seen as a powerful tool in the reestablishment of normal mitochondrial function. In line with this requirement, in this work and for the first time, a mitochondrial-targeting sequence (MTS) has been incorporated into previously researched peptides, to confer on them a targeting ability. These peptides were then considered to complex a plasmid DNA (pDNA) which contains the mitochondrial gene ND1 (mitochondrially encoded NADH dehydrogenase 1 protein), aiming at the formation of peptide-based nanoparticles. Currently, the ND1 plasmid is one of the most advanced bioengineered vectors for conducting research on mitochondrial gene expression. The formed complexes were characterized in terms of pDNA complexation capacity, morphology, size, surface charge and cytotoxic profile. These data revealed that the developed carriers possess suitable properties for pDNA delivery. Furthermore, in vitro studies illustrated the mitochondrial targeting ability of the novel peptide/pDNA complexes. A comparison between the different complexes revealed the most promising ones that complex pDNA and target mitochondria. This may contribute to the optimization of peptide-based non-viral systems to target mitochondria, instigating progress in mitochondrial gene therapy.
Collapse
Affiliation(s)
- Rúben Faria
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (R.F.); (A.S.)
| | - Eric Vivés
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34295 Montpellier, France; (E.V.); (P.B.)
| | - Prisca Boisguerin
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34295 Montpellier, France; (E.V.); (P.B.)
| | - Angela Sousa
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (R.F.); (A.S.)
| | - Diana Costa
- CICS-UBI—Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (R.F.); (A.S.)
| |
Collapse
|
38
|
Kumar R, Santa Chalarca CF, Bockman MR, Bruggen CV, Grimme CJ, Dalal RJ, Hanson MG, Hexum JK, Reineke TM. Polymeric Delivery of Therapeutic Nucleic Acids. Chem Rev 2021; 121:11527-11652. [PMID: 33939409 DOI: 10.1021/acs.chemrev.0c00997] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The advent of genome editing has transformed the therapeutic landscape for several debilitating diseases, and the clinical outlook for gene therapeutics has never been more promising. The therapeutic potential of nucleic acids has been limited by a reliance on engineered viral vectors for delivery. Chemically defined polymers can remediate technological, regulatory, and clinical challenges associated with viral modes of gene delivery. Because of their scalability, versatility, and exquisite tunability, polymers are ideal biomaterial platforms for delivering nucleic acid payloads efficiently while minimizing immune response and cellular toxicity. While polymeric gene delivery has progressed significantly in the past four decades, clinical translation of polymeric vehicles faces several formidable challenges. The aim of our Account is to illustrate diverse concepts in designing polymeric vectors towards meeting therapeutic goals of in vivo and ex vivo gene therapy. Here, we highlight several classes of polymers employed in gene delivery and summarize the recent work on understanding the contributions of chemical and architectural design parameters. We touch upon characterization methods used to visualize and understand events transpiring at the interfaces between polymer, nucleic acids, and the physiological environment. We conclude that interdisciplinary approaches and methodologies motivated by fundamental questions are key to designing high-performing polymeric vehicles for gene therapy.
Collapse
Affiliation(s)
- Ramya Kumar
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | - Matthew R Bockman
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Craig Van Bruggen
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christian J Grimme
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Joseph K Hexum
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
39
|
Recent advances in peptide-targeted micelleplexes: Current developments and future perspectives. Int J Pharm 2021; 597:120362. [PMID: 33556489 DOI: 10.1016/j.ijpharm.2021.120362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
The decoding of the human genome revolutionized the understanding of how genetics influence the interplay between health and disease, in a multidisciplinary perspective. Thus, the development of exogenous nucleic acids-based therapies has increased to overcome hereditary or acquired genetic-associated diseases. Gene drug delivery using non-viral systems, for instance micelleplexes, have been recognized as promising options for gene-target therapies. Micelleplexes are core-shell structures, at a nanometric scale, designed using amphiphilic block copolymers. These can self-assemble in an aqueous medium, leading to the formation of a hydrophilic and positively charged corona - that can transport nucleic acids, - and a hydrophobic core - which can transport poor water-soluble drugs. However, the performance of these types of carriers usually is hindered by several in vivo barriers. Fortunately, due to a significant amount of research, strategies to overcome these shortcomings emerged. With a wide range of structural features, good stability against proteolytic degradation, affordable characteristic, easy synthesis, low immunogenicity, among other advantages, peptides have increasingly gained popularity as target ligands for non-viral carriers. Hence, this review addresses the use of peptides with micelleplexes illustrating, through the analysis of in vitro and in vivo studies, the potential and future perspectives of this combination.
Collapse
|
40
|
Richter F, Mapfumo P, Martin L, Solomun JI, Hausig F, Frietsch JJ, Ernst T, Hoeppener S, Brendel JC, Traeger A. Improved gene delivery to K-562 leukemia cells by lipoic acid modified block copolymer micelles. J Nanobiotechnology 2021; 19:70. [PMID: 33676500 PMCID: PMC7936509 DOI: 10.1186/s12951-021-00801-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/09/2021] [Indexed: 12/23/2022] Open
Abstract
Although there has been substantial progress in the research field of gene delivery, there are some challenges remaining, e.g. there are still cell types such as primary cells and suspension cells (immune cells) known to be difficult to transfect. Cationic polymers have gained increasing attention due to their ability to bind, condense and mask genetic material, being amenable to scale up and highly variable in their composition. In addition, they can be combined with further monomers exhibiting desired biological and chemical properties, such as antioxidative, pH- and redox-responsive or biocompatible features. By introduction of hydrophobic monomers, in particular as block copolymers, cationic micelles can be formed possessing an improved chance of transfection in otherwise challenging cells. In this study, the antioxidant biomolecule lipoic acid, which can also be used as crosslinker, was incorporated into the hydrophobic block of a diblock copolymer, poly{[2-(dimethylamino)ethyl methacrylate]101-b-[n-(butyl methacrylate)124-co-(lipoic acid methacrylate)22]} (P(DMAEMA101-b-[nBMA124-co-LAMA22])), synthesized by RAFT polymerization and assembled into micelles (LAMA-mic). These micelles were investigated regarding their pDNA binding, cytotoxicity mechanisms and transfection efficiency in K-562 and HEK293T cells, the former representing a difficult to transfect, suspension leukemia cell line. The LAMA-mic exhibited low cytotoxicity at applied concentrations but demonstrated superior transfection efficiency in HEK293T and especially K-562 cells. In-depth studies on the transfection mechanism revealed that transfection efficiency in K-562 cells does not depend on the specific oncogenic fusion gene BCR-ABL alone. It is independent of the cellular uptake of polymer-pDNA complexes but correlates with the endosomal escape of the LAMA-mic. A comparison of the transfection efficiency of the LAMA-mic with structurally comparable micelles without lipoic acid showed that lipoic acid is not solely responsible for the superior transfection efficiency of the LAMA-mic. More likely, a synergistic effect of the antioxidative lipoic acid and the micellar architecture was identified. Therefore, the incorporation of lipoic acid into the core of hydrophobic-cationic micelles represents a promising tailor-made transfer strategy, which can potentially be beneficial for other difficult to transfect cell types.
Collapse
Affiliation(s)
- Friederike Richter
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Prosper Mapfumo
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Liam Martin
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jana I Solomun
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Franziska Hausig
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
| | - Jochen J Frietsch
- Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Thomas Ernst
- Klinik für Innere Medizin II, Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Stephanie Hoeppener
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Johannes C Brendel
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Anja Traeger
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstrasse 10, 07743, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany.
| |
Collapse
|
41
|
Lin M, Dai Y, Xia F, Zhang X. Advances in non-covalent crosslinked polymer micelles for biomedical applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 119:111626. [DOI: 10.1016/j.msec.2020.111626] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/08/2020] [Accepted: 10/10/2020] [Indexed: 12/26/2022]
|
42
|
Laurini E, Aulic S, Marson D, Fermeglia M, Pricl S. Cationic Dendrimers for siRNA Delivery: An Overview of Methods for In Vitro/In Vivo Characterization. Methods Mol Biol 2021; 2282:209-244. [PMID: 33928579 DOI: 10.1007/978-1-0716-1298-9_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This chapter reviews the different techniques for analyzing the chemical-physical properties, transfection efficiency, cytotoxicity, and stability of covalent cationic dendrimers (CCDs) and self-assembled cationic dendrons (ACDs) for siRNA delivery in the presence and absence of their nucleic cargos. On the basis of the reported examples, a standard essential set of techniques is described for each step of a siRNA/nanovector (NV) complex characterization process: (1) analysis of the basic chemical-physical properties of the NV per se; (2) characterization of the morphology, size, strength, and stability of the siRNA/NV ensemble; (3) characterization and quantification of the cellular uptake and release of the siRNA fragment; (4) in vitro and (5) in vivo experiments for the evaluation of the corresponding gene silencing activity; and (6) assessment of the intrinsic toxicity of the NV and the siRNA/NV complex.
Collapse
Affiliation(s)
- Erik Laurini
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTs), Department of Engineering and Architecture, University of Trieste, Trieste, Italy.
| | - Suzana Aulic
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTs), Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Domenico Marson
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTs), Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Maurizio Fermeglia
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTs), Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTs), Department of Engineering and Architecture, University of Trieste, Trieste, Italy
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| |
Collapse
|
43
|
Fuchs S, Ernst AU, Wang LH, Shariati K, Wang X, Liu Q, Ma M. Hydrogels in Emerging Technologies for Type 1 Diabetes. Chem Rev 2020; 121:11458-11526. [DOI: 10.1021/acs.chemrev.0c01062] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Stephanie Fuchs
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Alexander U. Ernst
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Long-Hai Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Kaavian Shariati
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Xi Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Qingsheng Liu
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Minglin Ma
- Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
44
|
Xiao F, Liu J, Zheng Y, Quan Z, Sun W, Fan Y, Luo C, Li H, Wu X. The targeted inhibition of prostate cancer by iron-based nanoparticles based on bioinformatics. J Biomater Appl 2020; 36:3-14. [PMID: 33283584 PMCID: PMC8217887 DOI: 10.1177/0885328220975249] [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: 12/24/2022]
Abstract
Prostate cancer is an epithelial malignant tumor of the prostate, and it is one of the malignant tumors with a high incidence of urogenital system in men. The local treatment of prostate cancer is mainly radical resection and radical radiotherapy, but they are not applicable to advanced prostate cancer. Systemic therapy mainly includes targeted therapy and immunotherapy which could cause many complications, and will affect the prognosis and quality of life of patients. It is urgent to find new treatments for prostate cancer. Bioinformatics offers hope for us to find reliable therapeutic targets. Bioinformatics can use the tumor informations in database and analyze them to screen out the best differentially expressed genes. Using the selected differentially expressed genes as targets, a gene interference plasmid was designed, and the constructed plasmid was used for targeted gene therapy. There are some problems about gene therapy that need to be solved, such as how to transfer genes to target cells is also an important challenge. Due to their large molecular weight and hydrophilic nature, they cannot enter cells through passive diffusion mechanisms. Here we synthesized a DNA carrier used surface modified iron based nanoparticles, and used it to load plasmid including ShRNA which can inhibit the expression of oncogene SLC4A4 selected by bioinformatics’ method. After that we use this iron based nanoparticles/plasmid DNA nanocomposite to treat prostate cancer cells in vitro and in vivo. The target gene SLC4A4 we had selected using bioinformatics had a strong effect on the proliferation of prostate cells; Our nanocomposite could inhibit the expression of SLC4A4 effectively, it had strong inhibitory effects on prostate cancer cells both in vivo and in vitro, and can be used as a potential method for prostate cancer treatment.
Collapse
Affiliation(s)
- Feng Xiao
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Jiayu Liu
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Yongbo Zheng
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Zhen Quan
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Wei Sun
- Fuling Center Hospital of Chongqing City, Chongqing, China
| | - Yao Fan
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Chunli Luo
- Chongqing Medical University, Chongqing, China
| | - Hailiang Li
- Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaohou Wu
- Department of Urology, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| |
Collapse
|
45
|
Madry H, Venkatesan JK, Carballo-Pedrares N, Rey-Rico A, Cucchiarini M. Scaffold-Mediated Gene Delivery for Osteochondral Repair. Pharmaceutics 2020; 12:pharmaceutics12100930. [PMID: 33003607 PMCID: PMC7601511 DOI: 10.3390/pharmaceutics12100930] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
Abstract
Osteochondral defects involve both the articular cartilage and the underlying subchondral bone. If left untreated, they may lead to osteoarthritis. Advanced biomaterial-guided delivery of gene vectors has recently emerged as an attractive therapeutic concept for osteochondral repair. The goal of this review is to provide an overview of the variety of biomaterials employed as nonviral or viral gene carriers for osteochondral repair approaches both in vitro and in vivo, including hydrogels, solid scaffolds, and hybrid materials. The data show that a site-specific delivery of therapeutic gene vectors in the context of acellular or cellular strategies allows for a spatial and temporal control of osteochondral neotissue composition in vitro. In vivo, implantation of acellular hydrogels loaded with nonviral or viral vectors has been reported to significantly improve osteochondral repair in translational defect models. These advances support the concept of scaffold-mediated gene delivery for osteochondral repair.
Collapse
Affiliation(s)
- Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany; (H.M.); (J.K.V.)
| | - Jagadeesh Kumar Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany; (H.M.); (J.K.V.)
| | - Natalia Carballo-Pedrares
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, S-15071 A Coruña, Spain; (N.C.-P.); (A.R.-R.)
| | - Ana Rey-Rico
- Cell Therapy and Regenerative Medicine Unit, Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña, S-15071 A Coruña, Spain; (N.C.-P.); (A.R.-R.)
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421 Homburg, Germany; (H.M.); (J.K.V.)
- Correspondence: ; Tel.: +49-684-1162-4987; Fax: +49-684-1162-4988
| |
Collapse
|
46
|
Tarvirdipour S, Huang X, Mihali V, Schoenenberger CA, Palivan CG. Peptide-Based Nanoassemblies in Gene Therapy and Diagnosis: Paving the Way for Clinical Application. Molecules 2020; 25:E3482. [PMID: 32751865 PMCID: PMC7435460 DOI: 10.3390/molecules25153482] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/26/2022] Open
Abstract
Nanotechnology approaches play an important role in developing novel and efficient carriers for biomedical applications. Peptides are particularly appealing to generate such nanocarriers because they can be rationally designed to serve as building blocks for self-assembling nanoscale structures with great potential as therapeutic or diagnostic delivery vehicles. In this review, we describe peptide-based nanoassemblies and highlight features that make them particularly attractive for the delivery of nucleic acids to host cells or improve the specificity and sensitivity of probes in diagnostic imaging. We outline the current state in the design of peptides and peptide-conjugates and the paradigms of their self-assembly into well-defined nanostructures, as well as the co-assembly of nucleic acids to form less structured nanoparticles. Various recent examples of engineered peptides and peptide-conjugates promoting self-assembly and providing the structures with wanted functionalities are presented. The advantages of peptides are not only their biocompatibility and biodegradability, but the possibility of sheer limitless combinations and modifications of amino acid residues to induce the assembly of modular, multiplexed delivery systems. Moreover, functions that nature encoded in peptides, such as their ability to target molecular recognition sites, can be emulated repeatedly in nanoassemblies. Finally, we present recent examples where self-assembled peptide-based assemblies with "smart" activity are used in vivo. Gene delivery and diagnostic imaging in mouse tumor models exemplify the great potential of peptide nanoassemblies for future clinical applications.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
- Department of Biosystem Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Xinan Huang
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
| | - Voichita Mihali
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
| | - Cora-Ann Schoenenberger
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
| | - Cornelia G. Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (X.H.); (V.M.)
| |
Collapse
|
47
|
Pereira-Silva M, Jarak I, Alvarez-Lorenzo C, Concheiro A, Santos AC, Veiga F, Figueiras A. Micelleplexes as nucleic acid delivery systems for cancer-targeted therapies. J Control Release 2020; 323:442-462. [DOI: 10.1016/j.jconrel.2020.04.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/09/2023]
|
48
|
Montero N, Alhajj MJ, Sierra M, Oñate-Garzon J, Yarce CJ, Salamanca CH. Development of Polyelectrolyte Complex Nanoparticles-PECNs Loaded with Ampicillin by Means of Polyelectrolyte Complexation and Ultra-High Pressure Homogenization (UHPH). Polymers (Basel) 2020; 12:E1168. [PMID: 32443668 PMCID: PMC7285317 DOI: 10.3390/polym12051168] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/09/2020] [Accepted: 05/14/2020] [Indexed: 12/27/2022] Open
Abstract
This study was focused on synthesizing, characterizing and evaluating the biological potential of Polyelectrolyte Complex Nanoparticles (PECNs) loaded with the antibiotic ampicillin. For this, the PECNs were produced initially by polyelectrolytic complexation (bottom-up method) and subsequently subjected to ultra-high pressure homogenization-UHPH (top-down method). The synthetic polymeric materials corresponding to the sodium salt of poly(maleic acid-alt-octadecene) (PAM-18Na) and the chloride salt of Eudragit E-100 (EuCl) were used, where the order of polyelectrolyte complexation, the polyelectrolyte ratio and the UHPH conditions on the PECNs features were evaluated. Likewise, PECNs were physicochemically characterized through particle size, polydispersity index, zeta potential, pH and encapsulation efficiency, whereas the antimicrobial effect was evaluated by means of the broth microdilution method employing ampicillin sensitive and resistant S. aureus strains. The results showed that the classical method of polyelectrolyte complexation (bottom-up) led to obtain polymeric complexes with large particle size and high polydispersity, where the 1:1 ratio between the titrant and receptor polyelectrolyte was the most critical condition. In contrast, the UHPH technique (top-down method) proved high performance to produce uniform polymeric complexes on the nanometric scale (particle size < 200 nm and PDI < 0.3). Finally, it was found there was a moderate increase in antimicrobial activity when ampicillin was loaded into the PECNs.
Collapse
Affiliation(s)
- Nicolle Montero
- Laboratorio de Diseño y Formulación de Productos Químicos y Derivados, Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Naturales, Universidad ICESI, Calle 18 No. 122-135, Cali 760035, Colombia; (N.M.); (M.J.A.); (M.S.); (C.J.Y.)
| | - Maria J. Alhajj
- Laboratorio de Diseño y Formulación de Productos Químicos y Derivados, Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Naturales, Universidad ICESI, Calle 18 No. 122-135, Cali 760035, Colombia; (N.M.); (M.J.A.); (M.S.); (C.J.Y.)
| | - Mariana Sierra
- Laboratorio de Diseño y Formulación de Productos Químicos y Derivados, Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Naturales, Universidad ICESI, Calle 18 No. 122-135, Cali 760035, Colombia; (N.M.); (M.J.A.); (M.S.); (C.J.Y.)
| | - Jose Oñate-Garzon
- Grupo de Investigación en Química y Biotecnología (QUIBIO), Facultad de Ciencias Básicas, Universidad Santiago de Cali, calle 5 No. 62-00, Cali 760035, Colombia;
| | - Cristhian J. Yarce
- Laboratorio de Diseño y Formulación de Productos Químicos y Derivados, Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Naturales, Universidad ICESI, Calle 18 No. 122-135, Cali 760035, Colombia; (N.M.); (M.J.A.); (M.S.); (C.J.Y.)
| | - Constain H. Salamanca
- Laboratorio de Diseño y Formulación de Productos Químicos y Derivados, Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Naturales, Universidad ICESI, Calle 18 No. 122-135, Cali 760035, Colombia; (N.M.); (M.J.A.); (M.S.); (C.J.Y.)
| |
Collapse
|
49
|
Interactions of the Lysosomotropic Detergent O-Methyl-Serine Dodecylamide Hydrochloride (MSDH) with Lipid Bilayer Membranes-Implications for Cell Toxicity. Int J Mol Sci 2020; 21:ijms21093136. [PMID: 32365555 PMCID: PMC7247706 DOI: 10.3390/ijms21093136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
O-methyl-serine dodecylamine hydrochloride (MSDH) is a detergent that accumulates selectively in lysosomes, a so-called lysosomotropic detergent, with unexpected chemical properties. At physiological pH, it spontaneously forms vesicles, which disassemble into small aggregates (probably micelles) below pH 6.4. In this study, we characterize the interaction between MSDH and liposomes at different pH and correlate the findings to toxicity in human fibroblasts. We find that the effect of MSDH on lipid membranes is highly pH-dependent. At neutral pH, the partitioning of MSDH into the liposome membrane is immediate and causes the leakage of small fluorophores, unless the ratio between MSDH and lipids is kept low. At pH 5, the partitioning of MSDH into the membrane is kinetically impeded since MSDH is charged and a high ratio between MSDH and the lipids is required to permeabilize the membrane. When transferred to cell culture conditions, the ratio between MSDH and plasma membrane lipids must therefore be low, at physiological pH, to maintain plasma membrane integrity. Transmission electron microscopy suggests that MSDH vesicles are taken up by endocytosis. As the pH of the endosomal compartment progressively drops, MSDH vesicles disassemble, leading to a high concentration of increasingly charged MSDH in small aggregates inside the lysosomes. At sufficiently high MSDH concentrations, the lysosome is permeabilized, the proteolytic content released to the cytosol and apoptotic cell death is induced.
Collapse
|
50
|
Hydrophilic/hydrophobic modifications of a PnBA-b-PDMAEA copolymer and complexation behaviour with short DNA. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.109636] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|