1
|
Karadurmus L, Erturk AS. Recent emerging trends in dendrimer research: Electrochemical sensors and their multifaceted applications in biomedical fields or healthcare. Biosens Bioelectron 2025; 273:117172. [PMID: 39823858 DOI: 10.1016/j.bios.2025.117172] [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: 12/14/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Dendrimers enhance the selectivity and sensitivity of sensors through their synthetic, highly branched, three-dimensional structures and large surface area. This unique architecture enables precise functionalization with various recognition elements, significantly improving the specificity and sensitivity of electrochemical sensors for detecting disease markers, biomolecules, and environmental pollutants. Dendrimer-based electrochemical sensors offer promising advancements in healthcare, such as detecting biomarkers for heart disease, monitoring blood glucose levels, and sensitively determining cancer-related proteins. Additionally, incorporating metals and conductive polymers into dendrimer nanocomposites can further enhance sensor performance. This review article provides a detailed overview of dendrimer's history, structure, properties, electrochemical properties, and synthesis methods. Particular attention has been paid to recent developments in the applications of dendrimers including electrochemical sensors, drug delivery, gene therapy and bioimaging. Recent progress in various applications of dendrimer-based electrochemical sensors developed over the last seven years, focusing on their healthcare applications and discussing the primary goals and challenges that will shape future research in this field, is also critically analyzed. These advances in dendrimer technology hold great potential for the development of novel therapeutics and expanded applications in sensor design.
Collapse
Affiliation(s)
- Leyla Karadurmus
- Department of Analytical Chemistry, Faculty of Pharmacy, Adiyaman University, Adiyaman, 02040, Türkiye
| | - Ali Serol Erturk
- Department of Analytical Chemistry, Faculty of Pharmacy, Adiyaman University, Adiyaman, 02040, Türkiye.
| |
Collapse
|
2
|
Xuan J, Wang Z, Huang Y, Liu Y, Han Y, Li M, Xiao M. DNA response element-based smart drug delivery systems for precise drug release. Biomater Sci 2024; 12:3550-3564. [PMID: 38832670 DOI: 10.1039/d4bm00138a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Smart drug delivery systems (DDSs) that respond to, interact with, or are actuated by biological signals or pathological abnormalities (e.g., the tumor microenvironment) for controllable drug release are appealing therapeutic platforms for cancer treatment. Owing to their inherent self-assembled nature, nucleic acids have emerged as programmable materials for the development of multifunctional structures. In response to external environmental stimuli, DNA response elements can serve as switches to trigger conformational changes in DNA structures. Their stimulus-responsive properties make them promising candidates for constructing smart DDSs, and advancements in DNA response element-based DDSs in the field of biomedicine have been made. This review summarizes different types of DNA response elements, including DNA aptamers, DNAzymes, disulfide bond-modified DNA, pH-responsive DNA motifs, and photocleavable DNA building blocks, and highlights the advancements in DNA response element-based smart DDSs for precise drug release. Finally, future challenges and perspectives in this field are discussed.
Collapse
Affiliation(s)
- Jinnan Xuan
- Hubei Key Laboratory of Photoelectric Materials and Devices, School of Materials Science and Engineering, Hubei Normal University, 11 Cihu Road, Huangshi 435002, P. R. China
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, P. R. China.
| | - Zhen Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Shaanxi Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P. R. China
| | - Yuting Huang
- Department of Radiotherapy, Chaohu Hospital of Anhui Medical University, 64 Chaohu North Road, Chaohu 238000, P. R. China
| | - Yisi Liu
- Hubei Key Laboratory of Photoelectric Materials and Devices, School of Materials Science and Engineering, Hubei Normal University, 11 Cihu Road, Huangshi 435002, P. R. China
| | - Yuqiang Han
- Hubei Key Laboratory of Photoelectric Materials and Devices, School of Materials Science and Engineering, Hubei Normal University, 11 Cihu Road, Huangshi 435002, P. R. China
| | - Man Li
- Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Shaanxi Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P. R. China
| | - Mingshu Xiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, P. R. China.
| |
Collapse
|
3
|
Bristow P, Schantz K, Moosbrugger Z, Martin K, Liebenberg H, Steimle S, Xiao Q, Percec V, Wilner SE. Aptamer-Targeted Dendrimersomes Assembled from Azido-Modified Janus Dendrimers "Clicked" to DNA. Biomacromolecules 2024; 25:1541-1549. [PMID: 38394608 PMCID: PMC10934268 DOI: 10.1021/acs.biomac.3c01108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
Amphiphilic Janus dendrimers (JDs), synthetic alternatives to lipids, have the potential to expand the scope of nanocarrier delivery systems. JDs self-assemble into vesicles called dendrimersomes, encapsulate both hydrophobic cargo and nucleic acids, and demonstrate enhanced stability in comparison to lipid nanoparticles (LNPs). Here, we report the ability to enhance the cellular uptake of Janus dendrimersomes using DNA aptamers. Azido-modified JDs were synthesized and conjugated to alkyne-modified DNAs using copper-catalyzed azide alkyne cycloaddition. DNA-functionalized JDs form nanometer-sized dendrimersomes in aqueous solution via thin film hydration. These vesicles, now displaying short DNAs, are then hybridized to transferrin receptor binding DNA aptamers. Aptamer-targeted dendrimersomes show improved cellular uptake as compared to control vesicles via fluorescence microscopy and flow cytometry. This work demonstrates the versatility of using click chemistry to conjugate functionalized JDs with biologically relevant molecules and the feasibility of targeting DNA-modified dendrimersomes for drug delivery applications.
Collapse
Affiliation(s)
- Paige Bristow
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Kyle Schantz
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Zoe Moosbrugger
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Kailey Martin
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Haley Liebenberg
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Stefan Steimle
- Department
of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19014, United States
| | - Qi Xiao
- Roy
& Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19014, United States
| | - Virgil Percec
- Roy
& Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19014, United States
| | - Samantha E. Wilner
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| |
Collapse
|
4
|
Hu R, Lan J, Zhang D, Shen W. Nanotherapeutics for prostate cancer treatment: A comprehensive review. Biomaterials 2024; 305:122469. [PMID: 38244344 DOI: 10.1016/j.biomaterials.2024.122469] [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: 09/27/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
Prostate cancer (PCa) is the most prevalent solid organ malignancy and seriously affects male health. The adverse effects of prostate cancer therapeutics can cause secondary damage to patients. Nanotherapeutics, which have special targeting abilities and controlled therapeutic release profiles, may serve as alternative agents for PCa treatment. At present, many nanotherapeutics have been developed to treat PCa and have shown better treatment effects in animals than traditional therapeutics. Although PCa nanotherapeutics are highly attractive, few successful cases have been reported in clinical practice. To help researchers design valuable nanotherapeutics for PCa treatment and avoid useless efforts, herein, we first reviewed the strategies and challenges involved in prostate cancer treatment. Subsequently, we presented a comprehensive review of nanotherapeutics for PCa treatment, including their targeting methods, controlled release strategies, therapeutic approaches and mechanisms. Finally, we proposed the future prospects of nanotherapeutics for PCa treatment.
Collapse
Affiliation(s)
- Ruimin Hu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jin Lan
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Dinglin Zhang
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Wenhao Shen
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
5
|
Klajnert-Maculewicz B, Janaszewska A, Majecka A. Dendrimersomes: Biomedical applications. Chem Commun (Camb) 2023; 59:14611-14625. [PMID: 37999927 DOI: 10.1039/d3cc03182a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
In recent years, dendrimer-based vesicles, known as dendrimersomes, have garnered significant attention as highly promising alternatives to lipid vesicles in a variety of biomedical applications. Dendrimersomes offer several advantages, including relatively straightforward synthesis, non-immunogenic properties, stability in circulation, and minimal size variability. These vesicles are composed of Janus dendrimers, which are polymers characterized by two dendritic wedges with different terminal groups - hydrophilic and hydrophobic. This dendrimer structure enables the self-assembly of dendrimersomes. The purpose of this highlight is to provide an overview of recent advancements achieved through the utilization of biomimetic dendrimersomes in various biomedical applications such as drug and nucleic acid delivery.
Collapse
Affiliation(s)
- Barbara Klajnert-Maculewicz
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Anna Janaszewska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland.
| | - Agata Majecka
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
6
|
Pranav, Laskar P, Jaggi M, Chauhan SC, Yallapu MM. Biomolecule-functionalized nanoformulations for prostate cancer theranostics. J Adv Res 2023; 51:197-217. [PMID: 36368516 PMCID: PMC10491979 DOI: 10.1016/j.jare.2022.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 10/21/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Even with the advancement in the areas of cancer nanotechnology, prostate cancer still poses a major threat to men's health. Nanomaterials and nanomaterial-derived theranostic systems have been explored for diagnosis, imaging, and therapy for different types of cancer still, for prostate cancer they have not delivered at full potential because of the limitations like in vivo biocompatibility, immune responses, precise targetability, and therapeutic outcome associated with the nanostructured system. AIM OF REVIEW Functionalizing nanomaterials with different biomolecules and bioactive agents provides advantages like specificity towards cancerous tumors, improved circulation time, and modulation of the immune response leading to early diagnosis and targeted delivery of cargo at the site of action. KEY SCIENTIFIC CONCEPTS OF REVIEW In this review, we have emphasized the classification and comparison of various nanomaterials based on biofunctionalization strategy and source of biomolecules such that it can be used for possible translation in clinical settings and future developments. This review highlighted the opportunities for embedding highly specific biological targeting moieties (antibody, aptamer, oligonucleotides, biopolymer, peptides, etc.) on nanoparticles which can improve the detection of prostate cancer-associated biomarkers at a very low limit of detection, direct visualization of prostate tumors and lastly for its therapy. Lastly, special emphasis was given to biomimetic nanomaterials which include functionalization with extracellular vesicles, exosomes and viral particles and their application for prostate cancer early detection and drug delivery. The present review paves a new pathway for next-generation biofunctionalized nanomaterials for prostate cancer theranostic application and their possibility in clinical translation.
Collapse
Affiliation(s)
- Pranav
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.
| |
Collapse
|
7
|
Ouyang X, Gao D, Shen J, Zhou Y, Gao Y, Lv Y, Wang Q, Yu G, Chu PK. Stimuli‐Responsive Dendritic Supramolecular Vector for Tumor‐Specific Gene Delivery. ADVANCED NANOBIOMED RESEARCH 2023; 3. [DOI: 10.1002/anbr.202200172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
To simplify the preparation of dendritic materials, host–guest molecular recognition and self‐assembly are utilized to form a supramolecular dendritic gene vector (DNCVP). DNCVP is constructed from an amino dendron‐conjugated naphthol, viologen containing pH‐sensitive hydrazone‐bond‐linked PEG, and CB[8] with a molar ratio of 1:1:1. The pH‐ and reducing‐sensitivity of DNCVP is verified, and the stimuli‐responsive capacity enables the vector tumor targeting gene delivery ability. Owing to the protection of surface PEG, the supramolecular engineering endows the delivery vector with low cytotoxicity and good biocompatibility that are confirmed by the MTT assay. The excellent delivery ability of genes is investigated by in vitro transfection of pEGFP, pGL3, and silencing of siGAPDH. In vivo studies demonstrate promoted tumor accumulation of genes mediated by the dual‐responsive DNCVP and the transfection efficiency at the tumor site is greatly improved benefiting from the dynamic nature of noncovalent interactions. This study reveals DNCVP is a promising supramolecular dendritic gene delivery vector, providing a sophisticated strategy for precise gene therapy.
Collapse
Affiliation(s)
- Xumei Ouyang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province School of Medicine Hangzhou City University Hangzhou 310015 China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment Zhuhai Institute of Translational Medicine Zhuhai People's Hospital Affiliated with Jinan University Jinan University Zhuhai Guangdong 519000 China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 China
| | - Dongruo Gao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province School of Medicine Hangzhou City University Hangzhou 310015 China
- College of Chemical and Biological Engineering Zhejiang University Zhejiang Hangzhou 310027 China
| | - Jie Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province School of Medicine Hangzhou City University Hangzhou 310015 China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 China
- Department of Physics Department of Materials Science and Engineering, and Department of Biomedical Engineering City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong China
| | - Yichen Zhou
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province School of Medicine Hangzhou City University Hangzhou 310015 China
| | - Ying Gao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province School of Medicine Hangzhou City University Hangzhou 310015 China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 China
| | - Yuanyuan Lv
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province School of Medicine Hangzhou City University Hangzhou 310015 China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences Zhejiang University Hangzhou 310058 China
- College of Chemical and Biological Engineering Zhejiang University Zhejiang Hangzhou 310027 China
| | - Qiwen Wang
- Department of Cardiology The First Affiliated Hospital School of Medicine Zhejiang University Hangzhou 310003 China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology Department of Chemistry Tsinghua University Beijing 100084 China
| | - Paul K. Chu
- Department of Physics Department of Materials Science and Engineering, and Department of Biomedical Engineering City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong China
| |
Collapse
|
8
|
Laskar P, Dhasmana A, Kotnala S, Jaggi M, Yallapu MM, Chauhan SC. Glutathione-Responsive Tannic Acid-Assisted FRET Nanomedicine for Cancer Therapy. Pharmaceutics 2023; 15:1326. [PMID: 37242568 PMCID: PMC10222396 DOI: 10.3390/pharmaceutics15051326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/28/2023] Open
Abstract
In cancer combination therapy, a multimodal delivery vector is used to improve the bioavailability of multiple anti-cancer hydrophobic drugs. Further, targeted delivery of therapeutics along with simultaneous monitoring of the drug release at the tumor site without normal organ toxicity is an emerging and effective strategy for cancer treatment. However, the lack of a smart nano-delivery system limits the application of this therapeutic strategy. To overcome this issue, a PEGylated dual drug, conjugated amphiphilic polymer (CPT-S-S-PEG-CUR), has been successfully synthesized by conjugating two hydrophobic fluorescent anti-cancer drugs, curcumin (CUR) and camptothecin (CPT), through an ester and a redox-sensitive disulfide (-S-S-) linkage, respectively, with a PEG chain via in situ two-step reactions. CPT-S-S-PEG-CUR is spontaneously self-assembled in the presence of tannic acid (TA, a physical crosslinker) into anionic, comparatively smaller-sized (~100 nm), stable nano-assemblies in water in comparison to only polymer due to stronger H-bond formation between polymer and TA. Further, due to the spectral overlap between CPT and CUR and a stable, smaller nano-assembly formation by the pro-drug polymer in water in presence of TA, a successful Fluorescence Resonance Energy Transfer (FRET) signal was generated between the conjugated CPT (FRET donor) and conjugated CUR (FRET acceptor). Interestingly, these stable nano-assemblies showed a preferential breakdown and release of CPT in a tumor-relevant redox environment (in the presence of 50 mM glutathione), leading to the disappearance of the FRET signal. These nano-assemblies exhibited a successful cellular uptake by the cancer cells and an enhanced antiproliferative effect in comparison to the individual drugs in cancer cells (AsPC1 and SW480). Such promising in vitro results with a novel redox-responsive, dual-drug conjugated, FRET pair-based nanosized multimodal delivery vector can be highly useful as an advanced theranostic system towards effective cancer treatment.
Collapse
Affiliation(s)
- Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- The Ångström Laboratory, Macromolecular Chemistry, Department of Chemistry, Uppsala University, 751 21 Uppsala, Sweden
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- Cancer Research Institute, Himalayan School of Biosciences, Swami Rama Himalayan University, Dehradun 248016, India
| | - Sudhir Kotnala
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Murali M. Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| |
Collapse
|
9
|
Shtykalova S, Deviatkin D, Freund S, Egorova A, Kiselev A. Non-Viral Carriers for Nucleic Acids Delivery: Fundamentals and Current Applications. Life (Basel) 2023; 13:903. [PMID: 37109432 PMCID: PMC10142071 DOI: 10.3390/life13040903] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Over the past decades, non-viral DNA and RNA delivery systems have been intensively studied as an alternative to viral vectors. Despite the most significant advantage over viruses, such as the lack of immunogenicity and cytotoxicity, the widespread use of non-viral carriers in clinical practice is still limited due to the insufficient efficacy associated with the difficulties of overcoming extracellular and intracellular barriers. Overcoming barriers by non-viral carriers is facilitated by their chemical structure, surface charge, as well as developed modifications. Currently, there are many different forms of non-viral carriers for various applications. This review aimed to summarize recent developments based on the essential requirements for non-viral carriers for gene therapy.
Collapse
Affiliation(s)
- Sofia Shtykalova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
- Faculty of Biology, Saint-Petersburg State University, Universitetskaya Embankment 7-9, 199034 Saint-Petersburg, Russia
| | - Dmitriy Deviatkin
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
- Faculty of Biology, Saint-Petersburg State University, Universitetskaya Embankment 7-9, 199034 Saint-Petersburg, Russia
| | - Svetlana Freund
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
- Faculty of Biology, Saint-Petersburg State University, Universitetskaya Embankment 7-9, 199034 Saint-Petersburg, Russia
| | - Anna Egorova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
| | - Anton Kiselev
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, Mendeleevskaya Line 3, 199034 Saint-Petersburg, Russia
| |
Collapse
|
10
|
Căta A, Ienașcu IMC, Ştefănuț MN, Roșu D, Pop OR. Properties and Bioapplications of Amphiphilic Janus Dendrimers: A Review. Pharmaceutics 2023; 15:589. [PMID: 36839911 PMCID: PMC9958631 DOI: 10.3390/pharmaceutics15020589] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Amphiphilic Janus dendrimers are arrangements containing both hydrophilic and hydrophobic units, capable of forming ordered aggregates by intermolecular noncovalent interactions between the dendrimer units. Compared to conventional dendrimers, these molecular self-assemblies possess particular and effective attributes i.e., the presence of different terminal groups, essential to design new elaborated materials. The present review will focus on the pharmaceutical and biomedical application of amphiphilic Janus dendrimers. Important information for the development of novel optimized pharmaceutical formulations, such as structural classification, synthetic pathways, properties and applications, will offer the complete characterization of this type of Janus dendrimers. This work will constitute an up-to-date background for dendrimer specialists involved in designing amphiphilic Janus dendrimer-based nanomaterials for future innovations in this promising field.
Collapse
Affiliation(s)
- Adina Căta
- National Institute of Research and Development for Electrochemistry and Condensed Matter, 144 Dr. A. P. Podeanu, 300569 Timişoara, Romania
| | - Ioana Maria Carmen Ienașcu
- National Institute of Research and Development for Electrochemistry and Condensed Matter, 144 Dr. A. P. Podeanu, 300569 Timişoara, Romania
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, “Vasile Goldiș” Western University of Arad, 86 Liviu Rebreanu, 310045 Arad, Romania
| | - Mariana Nela Ştefănuț
- National Institute of Research and Development for Electrochemistry and Condensed Matter, 144 Dr. A. P. Podeanu, 300569 Timişoara, Romania
| | - Dan Roșu
- National Institute of Research and Development for Electrochemistry and Condensed Matter, 144 Dr. A. P. Podeanu, 300569 Timişoara, Romania
| | - Oana-Raluca Pop
- Faculty of Pharmacy, University of Medicine and Pharmacy “Victor Babeș” Timișoara, 2 Eftimie Murgu Square, 300041 Timișoara, Romania
| |
Collapse
|
11
|
Sharma S, Mahajan SD, Chevli K, Schwartz SA, Aalinkeel R. Nanotherapeutic Approach to Delivery of Chemo- and Gene Therapy for Organ-Confined and Advanced Castration-Resistant Prostate Cancer. Crit Rev Ther Drug Carrier Syst 2023; 40:69-100. [PMID: 37075068 PMCID: PMC11007628 DOI: 10.1615/critrevtherdrugcarriersyst.2022043827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Treatments for late-stage prostate cancer (CaP) have not been very successful. Frequently, advanced CaP progresses to castration-resistant prostate cancer (CRPC), with 50#37;-70% of patients developing bone metastases. CaP with bone metastasis-associated clinical complications and treatment resistance presents major clinical challenges. Recent advances in the formulation of clinically applicable nanoparticles (NPs) have attracted attention in the fields of medicine and pharmacology with applications to cancer and infectious and neurological diseases. NPs have been rendered biocompatible, pose little to no toxicity to healthy cells and tissues, and are engineered to carry large therapeutic payloads, including chemo- and genetic therapies. Additionally, if required, targeting specificity can be achieved by chemically coupling aptamers, unique peptide ligands, or monoclonal antibodies to the surface of NPs. Encapsulating toxic drugs within NPs and delivering them specifically to their cellular targets overcomes the problem of systemic toxicity. Encapsulating highly labile genetic therapeutics such as RNA within NPs provides a protective environment for the payload during parenteral administration. The loading efficiencies of NPs have been maximized while the controlled their therapeutic cargos has been released. Theranostic ("treat and see") NPs have developed combining therapy with imaging capabilities to provide real-time, image-guided monitoring of the delivery of their therapeutic payloads. All of these NP accomplishments have been applied to the nanotherapy of late-stage CaP, offering a new opportunity for a previously dismal prognosis. This article gives an update on current developments in the use of nanotechnology for treating late-stage, castration-resistant CaP.
Collapse
Affiliation(s)
- Satish Sharma
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Supriya D. Mahajan
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Kent Chevli
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Stanley A. Schwartz
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Ravikumar Aalinkeel
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| |
Collapse
|
12
|
Aniu Lincy S, Allwin Richard Y, Vinitha T, Balamurugan K, Dharuman V. Streptavidin Fe 2O 3-gold nanoparticles functionalized theranostic liposome for antibiotic resistant bacteria and biotin sensing. Biosens Bioelectron 2023; 219:114849. [PMID: 36327565 DOI: 10.1016/j.bios.2022.114849] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/19/2022]
Abstract
Novel methods of sensing and treatment required to elicit potent humoral and cellular immune responses. Here, Streptavidin functionalized α-Fe2O3-Au nanoparticles (STV-Mag) loaded cationic carbomate cholesterol is used as a carrier to release antibacterial thymol drug for Staphylococcus aureus (S. aureus) infected Caenorhabditis elegans (C. elegans). Pertaining to theranostic applications, efficient antimicrobial activity, and non-stimulated drug release and biotin dependent S. aureus growth were studied in-vivo. While STV-Mag was tethered on mercaptobenzoic acid (MBA) molecular cushion for label free streptavidin-biotin electrochemical sensing, the STV-Mag-carbomate cholesterol (STV-Mag-cCHOL liposome) vesicle with loaded drug was tethered on MBA for non-stimulant drug release through specific cholesterol-S. aureus interaction and confirmed electrochemically. Selectivity was confirmed using other pathogens, E. coli, Proteus and Enterococcus bacterium through antimicrobial studies along with S. aureus. The biotin sensing showed linear range from 10-15 to 10-3 M, which was not obtained by conventional methods. Fourier-Transform Infra-red (FT-IR), X-ray diffraction (XRD), Scanning Electron Microscopy (SEM) and Transmission Electron Microscopy (TEM) techniques were used to characterize the nanoparticulate system.
Collapse
Affiliation(s)
- Sebastinbaskar Aniu Lincy
- Molecular Electronics Laboratory, Department of Bioelectronics and Biosensors, Science Campus, Alagappa University, Karaikudi, 630003, Tamilnadu, India
| | - Yesurajan Allwin Richard
- Molecular Electronics Laboratory, Department of Bioelectronics and Biosensors, Science Campus, Alagappa University, Karaikudi, 630003, Tamilnadu, India
| | - Thondimuthu Vinitha
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, 630003, Tamilnadu, India
| | - Krishnaswamy Balamurugan
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, 630003, Tamilnadu, India
| | - Venkataraman Dharuman
- Molecular Electronics Laboratory, Department of Bioelectronics and Biosensors, Science Campus, Alagappa University, Karaikudi, 630003, Tamilnadu, India.
| |
Collapse
|
13
|
Wang T, Qin J, Cheng J, Li C, Du J. Intelligent design of polymersomes for antibacterial and anticancer applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1822. [PMID: 35673991 DOI: 10.1002/wnan.1822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 01/25/2023]
Abstract
Polymersomes (or polymer vesicles) have attracted much attention for biomedical applications in recent years because their lumen can be used for drug delivery and their coronas and membrane can be modified with a variety of functional groups. Thus, polymersomes are very suitable for improved antibacterial and anticancer therapy. This review mainly highlighted recent advances in the synthetic protocols and design principles of intelligent antibacterial and anticancer polymersomes. Antibacterial polymersomes are divided into three categories: polymersomes as antibiotic nanocarriers, intrinsically antibacterial polymersomes, and antibacterial polymersomes with supplementary means including photothermal and photodynamic therapy. Similarly, the anticancer polymersomes are divided into two categories: polymersomes-based delivery systems and anticancer polymersomes with supplementary means. In addition, the bilateral relationship between bacteria and cancer is addressed, since more and more evidences show that bacteria may cause cancer or promote cancer progression. Finally, prospective on next-generation antibacterial and anticancer polymersomes are discussed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Tao Wang
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China
| | - Jinlong Qin
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China.,Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiajing Cheng
- Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Li
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, China.,Department of Gynecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
14
|
Wang Z, Song W, Sheng R, Guo X, Hao L, Zhang X. Controlled preparation of cholesterol-bearing polycations with pendent l-lysine for efficient gene delivery. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2058943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Zhao Wang
- School of Material Engineering, Jinling Institute of Technology, Nanjing, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing, China
| | - Wenli Song
- School of Material Engineering, Jinling Institute of Technology, Nanjing, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing, China
| | - Ruilong Sheng
- CQM-Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyu Guo
- School of Material Engineering, Jinling Institute of Technology, Nanjing, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing, China
| | - Lingyun Hao
- School of Material Engineering, Jinling Institute of Technology, Nanjing, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing, China
| | - Xiaojuan Zhang
- School of Material Engineering, Jinling Institute of Technology, Nanjing, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Nanjing, China
| |
Collapse
|
15
|
Sztandera K, Gorzkiewicz M, Bątal M, Arkhipova V, Knauer N, Sánchez-Nieves J, de la Mata FJ, Gómez R, Apartsin E, Klajnert-Maculewicz B. Triazine–Carbosilane Dendrimersomes Enhance Cellular Uptake and Phototoxic Activity of Rose Bengal in Basal Cell Skin Carcinoma Cells. Int J Nanomedicine 2022; 17:1139-1154. [PMID: 35321027 PMCID: PMC8935628 DOI: 10.2147/ijn.s352349] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/14/2022] [Indexed: 12/11/2022] Open
Abstract
Background The search for new formulations for photodynamic therapy is intended to improve the outcome of skin cancer treatment using significantly reduced doses of photosensitizer, thereby avoiding side effects. The incorporation of photosensitizers into nanoassemblies is a versatile way to increase the efficiency and specificity of drug delivery into target cells. Herein, we report the loading of rose bengal into vesicle-like constructs of amphiphilic triazine-carbosilane dendrons (dendrimersomes) as well as biophysical and in vitro characterization of this novel nanosystem. Methods Using established protocol and analytical and spectroscopy techniques we were able to synthesized dendrons with strictly designed properties. Engaging biophysical methods (hydrodynamic diameter and zeta potential measurements, analysis of spectral properties, transmission electron microscopy) we confirmed assembling of our nanosystem. A set of in vitro techniques was used for determination ROS generation, (ABDA and H2DCFDA probes), cell viability (MTT assay) and cellular uptake (flow cytometry and confocal microscopy). Results Encapsulation of rose bengal inside dendrimersomes enhances cellular uptake, intracellular ROS production and concequently, the phototoxicity of this photosensitizer. Conclusion Triazine-carbosilane dendrimersomes show high capacity as drug carriers for anticancer photodynamic therapy.
Collapse
Affiliation(s)
- Krzysztof Sztandera
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, 90-236, Poland
| | - Michał Gorzkiewicz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, 90-236, Poland
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | - Mateusz Bątal
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, 90-236, Poland
| | - Valeria Arkhipova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, 630090, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Nadezhda Knauer
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, 630090, Russia
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, 630099, Russia
- Clinic for Neurosurgery, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | - Javier Sánchez-Nieves
- Departamento de Química Orgánica y Química Inorgánica, UAH-IQAR, Alcalá de Henares, 28805, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, 28029, Spain
| | - Fco Javier de la Mata
- Departamento de Química Orgánica y Química Inorgánica, UAH-IQAR, Alcalá de Henares, 28805, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, 28029, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Madrid, 28034, Spain
| | - Rafael Gómez
- Departamento de Química Orgánica y Química Inorgánica, UAH-IQAR, Alcalá de Henares, 28805, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, 28029, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Madrid, 28034, Spain
| | - Evgeny Apartsin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, 630090, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, 630090, Russia
- Laboratoire de Chimie de Coordination CNRS, Toulouse, 31077, France
| | - Barbara Klajnert-Maculewicz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, 90-236, Poland
- Correspondence: Barbara Klajnert-Maculewicz, Department of General Biophysics, Pomorska 141/143, Łódź, 90-236, Poland, Tel +48 42 635 44 29, Fax +48 42 635 4474, Email
| |
Collapse
|
16
|
Meewan J, Somani S, Laskar P, Irving C, Mullin M, Woods S, Roberts CW, Alzahrani AR, Ferro VA, McGill S, Weidt S, Burchmore R, Dufès C. Limited Impact of the Protein Corona on the Cellular Uptake of PEGylated Zein Micelles by Melanoma Cancer Cells. Pharmaceutics 2022; 14:pharmaceutics14020439. [PMID: 35214171 PMCID: PMC8877401 DOI: 10.3390/pharmaceutics14020439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 12/17/2022] Open
Abstract
The formation of a protein layer “corona” on the nanoparticle surface upon entry into a biological environment was shown to strongly influence the interactions with cells, especially affecting the uptake of nanomedicines. In this work, we present the impact of the protein corona on the uptake of PEGylated zein micelles by cancer cells, macrophages, and dendritic cells. Zein was successfully conjugated with poly(ethylene glycol) (PEG) of varying chain lengths (5K and 10K) and assembled into micelles. Our results demonstrate that PEGylation conferred stealth effects to the zein micelles. The presence of human plasma did not impact the uptake levels of the micelles by melanoma cancer cells, regardless of the PEG chain length used. In contrast, it decreased the uptake by macrophages and dendritic cells. These results therefore make PEGylated zein micelles promising as potential drug delivery systems for cancer therapy.
Collapse
Affiliation(s)
- Jitkasem Meewan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Sukrut Somani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Partha Laskar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
- Department of Immunology and Microbiology, University of Texas Health Rio Grande Valley, 5300 North L Street 881 Madison, McAllen, TX 78504, USA
| | - Craig Irving
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK;
| | - Margaret Mullin
- Glasgow Imaging Facility, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK;
| | - Stuart Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Craig W. Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Abdullah R. Alzahrani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
- Department of Pharmacology & Toxicology, Faculty of Medicine, Umm Al-Qura University, Al-Abidiyah, P.O. Box 13578, Makkah 21955, Saudi Arabia
| | - Valerie A. Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
| | - Suzanne McGill
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; (S.M.); (S.W.); (R.B.)
| | - Stefan Weidt
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; (S.M.); (S.W.); (R.B.)
| | - Richard Burchmore
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; (S.M.); (S.W.); (R.B.)
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (J.M.); (S.S.); (P.L.); (S.W.); (C.W.R.); (A.R.A.); (V.A.F.)
- Correspondence: ; Tel.: +44-141-548-3796
| |
Collapse
|
17
|
|
18
|
Laskar P, Dufès C. Emergence of cationic polyamine dendrimersomes: design, stimuli sensitivity and potential biomedical applications. NANOSCALE ADVANCES 2021; 3:6007-6026. [PMID: 34765868 PMCID: PMC8548884 DOI: 10.1039/d1na00536g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/30/2021] [Indexed: 06/01/2023]
Abstract
For decades, self-assembled lipid vesicles have been widely used in clinics as nanoscale delivery systems for various biomedical applications, including treatment of various diseases. Due to their core-shell architecture and versatile nature, they have been successfully used as carriers for the delivery of a wide range of therapeutic cargos, including drugs and nucleic acids, in cancer treatment. Recently, surface-modified polyamine dendrimer-based vesicles, or dendrimersomes, have emerged as promising alternatives to lipid vesicles for various biomedical applications, due to their ease of synthesis, non-immunogenicity, stability in circulation and lower size polydispersity. This mini-review provides an overview of the recent advances resulting from the use of biomimetic hydrophobically-modified polyamine-based dendrimersomes towards biomedical applications, focusing mainly on the two most widely used polyamine dendrimers, namely polyamidoamine (PAMAM) and poly(propylene imine) (PPI) dendrimers.
Collapse
Affiliation(s)
- Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley McAllen TX 78504 USA
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde 161 Cathedral Street Glasgow G4 0RE UK
| |
Collapse
|
19
|
Niu B, Liao K, Zhou Y, Wen T, Quan G, Pan X, Wu C. Application of glutathione depletion in cancer therapy: Enhanced ROS-based therapy, ferroptosis, and chemotherapy. Biomaterials 2021; 277:121110. [PMID: 34482088 DOI: 10.1016/j.biomaterials.2021.121110] [Citation(s) in RCA: 562] [Impact Index Per Article: 140.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 01/17/2023]
Abstract
Glutathione (GSH) is an important member of cellular antioxidative system. In cancer cells, a high level of GSH is indispensable to scavenge excessive reactive oxygen species (ROS) and detoxify xenobiotics, which make it a potential target for cancer therapy. Plenty of studies have shown that loss of intracellular GSH makes cancer cells more susceptible to oxidative stress and chemotherapeutic agents. GSH depletion has been proved to improve the therapeutic efficacy of ROS-based therapy (photodynamic therapy, sonodynamic therapy, and chemodynamic therapy), ferroptosis, and chemotherapy. In this review, various strategies for GSH depletion used in cancer therapy are comprehensively summarized and discussed. First, the functions of GSH in cancer cells are analyzed to elucidate the necessity of GSH depletion in cancer therapy. Then, the synthesis and metabolism of GSH are briefly introduced to bring up some crucial targets for GSH modulation. Finally, different approaches to GSH depletion in the literature are classified and discussed in detail according to their mechanisms. Particularly, functional materials with GSH-consuming ability based on nanotechnology are elaborated due to their unique advantages and potentials. This review presents the ingenious application of GSH-depleting strategy in cancer therapy for improving the outcomes of various therapeutic regimens, which may provide useful guidance for designing intelligent drug delivery system.
Collapse
Affiliation(s)
- Boyi Niu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Kaixin Liao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yixian Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ting Wen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Chuanbin Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
20
|
Sharma D, Arora S, Singh J, Layek B. A review of the tortuous path of nonviral gene delivery and recent progress. Int J Biol Macromol 2021; 183:2055-2073. [PMID: 34087309 PMCID: PMC8266766 DOI: 10.1016/j.ijbiomac.2021.05.192] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Gene therapy encompasses the transfer of exogenous genetic materials into the patient's target cells to treat or prevent diseases. Nevertheless, the transfer of genetic material into desired cells is challenging and often requires specialized tools or delivery systems. For the past 40 years, scientists are mainly pursuing various viruses as gene delivery vectors, and the overall progress has been slow and far from the expectation. As an alternative, nonviral vectors have gained substantial attention due to their several advantages, including superior safety profile, enhanced payload capacity, and stealth abilities. Since nonviral vectors encounter multiple extra- and intra-cellular barriers limiting the transfer of genetic payload into the target cell nucleus, we have discussed these barriers in detail for this review. A direct approach, utilizing physical methods like electroporation, sonoporation, gene gun, eliminate the requirement for a specific carrier for gene delivery. In contrast, chemical methods of gene transfer exploit natural or synthetic compounds as carriers to increase cellular targeting and gene therapy effectiveness. We have also emphasized the recent advancements aimed at enhancing the current nonviral approaches. Therefore, in this review, we have focused on discussing the current evolving state of nonviral gene delivery systems and their future perspectives.
Collapse
Affiliation(s)
- Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo 58105, ND, USA.
| |
Collapse
|
21
|
Yu C, Li L, Hu P, Yang Y, Wei W, Deng X, Wang L, Tay FR, Ma J. Recent Advances in Stimulus-Responsive Nanocarriers for Gene Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2100540. [PMID: 34306980 PMCID: PMC8292848 DOI: 10.1002/advs.202100540] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/07/2021] [Indexed: 05/29/2023]
Abstract
Gene therapy provides a promising strategy for curing monogenetic disorders and complex diseases. However, there are challenges associated with the use of viral delivery vectors. The advent of nanomedicine represents a quantum leap in the application of gene therapy. Recent advances in stimulus-responsive nonviral nanocarriers indicate that they are efficient delivery systems for loading and unloading of therapeutic nucleic acids. Some nanocarriers are responsive to cues derived from the internal environment, such as changes in pH, redox potential, enzyme activity, reactive oxygen species, adenosine triphosphate, and hypoxia. Others are responsive to external stimulations, including temperature gradients, light irradiation, ultrasonic energy, and magnetic field. Multiple stimuli-responsive strategies have also been investigated recently for experimental gene therapy.
Collapse
Affiliation(s)
- Cheng Yu
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Long Li
- Department of OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Pei Hu
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Yan Yang
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Wei Wei
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Xin Deng
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | - Lu Wang
- Department of OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| | | | - Jingzhi Ma
- Department of StomatologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei Province430030China
| |
Collapse
|
22
|
Wang Z, Sun J, Li M, Luo T, Shen Y, Cao A, Sheng R. Natural steroid-based cationic copolymers cholesterol/diosgenin- r-PDMAEMAs and their pDNA nanoplexes: impact of steroid structures and hydrophobic/hydrophilic ratios on pDNA delivery. RSC Adv 2021; 11:19450-19460. [PMID: 35479247 PMCID: PMC9033666 DOI: 10.1039/d1ra00223f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/05/2021] [Indexed: 11/21/2022] Open
Abstract
Using natural-based lipids to construct biocompatible, controllable and efficient nanocarriers and elucidating their structure-function relationships, was regarded as an important area for creating sustainable biomaterials. Herein, we utilized two natural steroids: cholesterol and diosgenin (bearing different hydrophobic tails) as the building blocks, to synthesize a series of natural steroid-based cationic random copolymers PMA6Chol-r-PDMAEMA and PMA6Dios-r-PDMAEMA via RAFT polymerization. The results demonstrated that the steroid-r-PDMAEMA copolymers could efficiently bind pDNA (N/P < 3.0) and then form near-spherical shape (142-449 nm) and positively-charged (+11.5 to +19.6 mV) nanoparticles. The in vitro cytotoxicity and gene transfection efficiency greatly depend on the steroid hydrophobic tail structures and steroid/PDMAEMA block ratios. Optimum transfection efficiency of the (Chol-P1/pDNA and Dios-P3/pDNA) nanoplexes could reach to 18.1-31.2% of the PEI-25K/pDNA complex. Moreover, all of the steroid-r-PDMAEMA/Cy3-pDNA nanoplexes have an obvious "lysosome localization" effect, indicating the steroid structures do not remarkably influence the intracellular localization behaviors of these nanoplexes.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai 200072 China.,School of Material Engineering, Jinling Institute of Technology Nanjing 211169 China.,CAS Key Laboratory of Synthetic and Self-assembly Chemistry for Organic Functional Molecules, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences 345 Lingling Road Shanghai 200032 China
| | - Jingjing Sun
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai 200072 China.,CAS Key Laboratory of Synthetic and Self-assembly Chemistry for Organic Functional Molecules, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences 345 Lingling Road Shanghai 200032 China
| | - Mingrui Li
- CAS Key Laboratory of Synthetic and Self-assembly Chemistry for Organic Functional Molecules, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences 345 Lingling Road Shanghai 200032 China
| | - Ting Luo
- CAS Key Laboratory of Synthetic and Self-assembly Chemistry for Organic Functional Molecules, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences 345 Lingling Road Shanghai 200032 China
| | - Yulin Shen
- School of Material Engineering, Jinling Institute of Technology Nanjing 211169 China
| | - Amin Cao
- CAS Key Laboratory of Synthetic and Self-assembly Chemistry for Organic Functional Molecules, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences 345 Lingling Road Shanghai 200032 China
| | - Ruilong Sheng
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University Shanghai 200072 China.,CAS Key Laboratory of Synthetic and Self-assembly Chemistry for Organic Functional Molecules, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences 345 Lingling Road Shanghai 200032 China.,CQM-Centro de Quimica da Madeira, Universidade da Madeira Campus da Penteada Funchal Madeira 9000-390 Portugal
| |
Collapse
|
23
|
Cell membrane cloaked nanomedicines for bio-imaging and immunotherapy of cancer: Improved pharmacokinetics, cell internalization and anticancer efficacy. J Control Release 2021; 335:130-157. [PMID: 34015400 DOI: 10.1016/j.jconrel.2021.05.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/13/2023]
Abstract
Despite enormous advancements in the field of oncology, the innocuous and effectual treatment of various types of malignancies remained a colossal challenge. The conventional modalities such as chemotherapy, radiotherapy, and surgery have been remained the most viable options for cancer treatment, but lacking of target-specificity, optimum safety and efficacy, and pharmacokinetic disparities are their impliable shortcomings. Though, in recent decades, numerous encroachments in the field of onco-targeted drug delivery have been adapted but several limitations (i.e., short plasma half-life, early clearance by reticuloendothelial system, immunogenicity, inadequate internalization and localization into the onco-tissues, chemoresistance, and deficient therapeutic efficacy) associated with these onco-targeted delivery systems limits their clinical viability. To abolish the aforementioned inadequacies, a promising approach has been emerged in which stealthing of synthetic nanocarriers has been attained by cloaking them into the natural cell membranes. These biomimetic nanomedicines not only retain characteristics features of the synthetic nanocarriers but also inherit the cell-membrane intrinsic functionalities. In this review, we have summarized preparation methods, mechanism of cloaking, and pharmaceutical and therapeutic superiority of cell-membrane camouflaged nanomedicines in improving the bio-imaging and immunotherapy against various types of malignancies. These pliable adaptations have revolutionized the current drug delivery strategies by optimizing the plasma circulation time, improving the permeation into the cancerous microenvironment, escaping the immune evasion and rapid clearance from the systemic circulation, minimizing the immunogenicity, and enabling the cell-cell communication via cell membrane markers of biomimetic nanomedicines. Moreover, the preeminence of cell-membrane cloaked nanomedicines in improving the bio-imaging and theranostic applications, alone or in combination with phototherapy or radiotherapy, have also been pondered.
Collapse
|
24
|
Laskar P, Somani S, Mullin M, Tate RJ, Warzecha M, Bowering D, Keating P, Irving C, Leung HY, Dufès C. Octadecyl chain-bearing PEGylated poly(propyleneimine)-based dendrimersomes: physicochemical studies, redox-responsiveness, DNA condensation, cytotoxicity and gene delivery to cancer cells. Biomater Sci 2021; 9:1431-1448. [PMID: 33404026 DOI: 10.1039/d0bm01441a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
Stimuli-responsive nanocarriers have become increasingly important for nucleic acid and drug delivery in cancer therapy. Here, we report the synthesis, characterization and evaluation of disulphide-linked, octadecyl (C18 alkyl) chain-bearing PEGylated generation 3-diaminobutyric polypropylenimine dendrimer-based vesicles (or dendrimersomes) for gene delivery. The lipid-bearing PEGylated dendrimer was successfully synthesized through in situ two-step reaction. It was able to spontaneously self-assemble into stable, cationic, nanosized vesicles, with low critical aggregation concentration value, and also showed redox-responsiveness in presence of a glutathione concentration similar to that of the cytosolic reducing environment. In addition, it was able to condense more than 70% of DNA at dendrimer: DNA weight ratios of 5 : 1 and higher. This dendriplex resulted in an enhanced cellular uptake of DNA at dendrimer: DNA weight ratios of 10 : 1 and 20 : 1, by up to 16-fold and by up to 28-fold compared with naked DNA in PC-3 and DU145 prostate cancer cell lines respectively. At a dendrimer: DNA weight ratio of 20 : 1, it led to an increase in gene expression in PC-3 and DU145 cells, compared with DAB dendriplex. These octadecyl chain-bearing, PEGylated dendrimer-based vesicles are therefore promising redox-sensitive drug and gene delivery systems for potential applications in combination cancer therapy.
Collapse
Affiliation(s)
- Partha Laskar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mollazadeh S, Mackiewicz M, Yazdimamaghani M. Recent advances in the redox-responsive drug delivery nanoplatforms: A chemical structure and physical property perspective. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 118:111536. [PMID: 33255089 DOI: 10.1016/j.msec.2020.111536] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
Poor water solubility, off-target toxicity, and small therapeutic window are among major obstacles for the development of drug products. Redox-responsive drug delivery nanoplatforms not only overcome the delivery and pharmacokinetic pitfalls observed in conventional drug delivery, but also leverage the site-specific delivery properties. Cleavable diselenide and disulfide bonds in the presence of elevated reactive oxygen species (ROS) and glutathione concentration are among widely used stimuli-responsive bonds to design nanocarriers. This review covers a wide range of redox-responsive chemical structures and their properties for designing nanoparticles aiming controlled loading, delivery, and release of hydrophobic anticancer drugs at tumor site.
Collapse
Affiliation(s)
- Shirin Mollazadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marcin Mackiewicz
- Faculty of Chemistry, Biological and Chemical Research Center, University of Warsaw, Poland
| | - Mostafa Yazdimamaghani
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
26
|
Lipodendriplexes mediated enhanced gene delivery: a cellular to pre-clinical investigation. Sci Rep 2020; 10:21446. [PMID: 33293580 PMCID: PMC7723038 DOI: 10.1038/s41598-020-78123-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/27/2020] [Indexed: 01/10/2023] Open
Abstract
Clinical success of effective gene therapy is mainly hampered by the insufficiency of safe and efficient internalization of a transgene to the targeted cellular site. Therefore, the development of a safe and efficient nanocarrier system is one of the fundamental challenges to transfer the therapeutic genes to the diseased cells. Polyamidoamine (PAMAM) dendrimer has been used as an efficient non-viral gene vector (dendriplexes) but the toxicity and unusual biodistribution induced by the terminal amino groups (–NH2) limit its in vivo applications. Hence, a state of the art lipid modification with PAMAM based gene carrier (lipodendriplexes) was planned to investigate theirs in vitro (2D and 3D cell culture) and in vivo behaviour. In vitro pDNA transfection, lactate dehydrogenase (LDH) release, reactive oxygen species (ROS) generation, cellular protein contents, live/dead staining and apoptosis were studied in 2D cell culture of HEK-293 cells while GFP transfection, 3D cell viability and live/dead staining of spheroids were performed in its 3D cell culture. Acute toxicity studies including organ to body index ratio, hematological parameters, serum biochemistry, histopathological profiles and in vivo transgene expression were assessed in female BALB/c mice. The results suggested that, in comparison to dendriplexes the lipodendriplexes exhibited significant improvement of pDNA transfection (p < 0.001) with lower LDH release (p < 0.01) and ROS generation (p < 0.05). A substantially higher cellular protein content (p < 0.01) and cell viability were also observed in 2D culture. A strong GFP expression with an improved cell viability profile (p < 0.05) was indicated in lipodendriplexes treated 3D spheroids. In vivo archives showed the superiority of lipid-modified nanocarrier system, depicted a significant increase in green fluorescent protein (GFP) expression in the lungs (p < 0.01), heart (p < 0.001), liver (p < 0.001) and kidneys (p < 0.001) with improved serum biochemistry and hematological profile as compared to unmodified dendriplexes. No tissue necrosis was evident in the animal groups treated with lipid-shielded molecules. Therefore, a non-covalent conjugation of lipids with PAMAM based carrier system could be considered as a promising approach for an efficient and biocompatible gene delivery system.
Collapse
|
27
|
Zheng Y, Weng C, Cheng C, Zhao J, Yang R, Zhang Q, Ding M, Tan H, Fu Q. Multiblock Copolymers toward Segmentation-Driven Morphological Transition. Macromolecules 2020. [DOI: 10.1021/acs.macromol.0c00374] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Yi Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Chuang Weng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Cheng Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Jinling Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Rui Yang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Qin Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Mingming Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Qiang Fu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
28
|
Chen H, Fan X, Zhao Y, Zhi D, Cui S, Zhang E, Lan H, Du J, Zhang Z, Zhang S, Zhen Y. Stimuli-Responsive Polysaccharide Enveloped Liposome for Targeting and Penetrating Delivery of survivin-shRNA into Breast Tumor. ACS APPLIED MATERIALS & INTERFACES 2020; 12:22074-22087. [PMID: 32083833 DOI: 10.1021/acsami.9b22440] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Silencing the inhibitor of apoptosis (IAP) by RNAi is a promising method for tumor therapy. One of the major challenges lies in how to sequentially overcome the system barriers in the course of the tumor targeting delivery, especially in the tumor accumulation and penetration. Herein we developed a novel stimuli-responsive polysaccharide enveloped liposome carrier, which was constructed by layer-by-layer depositing redox-sensitive amphiphilic chitosan (CS) and hyaluronic acid (HA) onto the liposome and then loading IAP inhibitor survivin-shRNA gene and permeation promoter hyaluronidase (HAase) sequentially. The as-prepared HA/HAase/CS/liposome/shRNA (HCLR) nanocarrier was verified to be stable in blood circulation due to the negative charged HA shield. The tumor targeting recognition and the enhanced tumor accumulation of HCLR were visualized by fluorescence resonance energy transfer (FRET) and in vivo fluorescence biodistribution. The deshielding of HA and the protonizing of CS in slightly acidic tumor extracellular pH environment (pHe, 6.8-6.5) were demonstrated by ζ potential change from -23.1 to 29.9 mV. The pHe-responsive HAase release was confirmed in the tumor extracellular mimicking environments, and the intratumoral biodistribution showed that the tumor penetration of HCLR was improved. The cell uptake of HCLR in pHe environment was significantly enhanced compared with that in physiological pH environment. The increased shRNA release of HCLR was approved in 10 mM glutathione (GSH) and tumor cells. Surprisingly, HCLR suppressed the tumor growth markedly through survivin silencing and meanwhile maintained low toxicity to mice. This study indicates that the novel polysaccharide enveloped HCLR is promising in clinical translation, thanks to the stimuli-triggered tumor accumulation, tumor penetration, cell uptake, and intracellular gene release.
Collapse
Affiliation(s)
- Huiying Chen
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, 18 Liaohe West Road, Dalian 116600, Liaoning Province People's Republic of China
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Ganjingzi District, Dalian 116024, Liaoning Province People's Republic of China
| | - Xuefeng Fan
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, 18 Liaohe West Road, Dalian 116600, Liaoning Province People's Republic of China
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, 18 Liaohe West Road, Dalian 116600, Liaoning Province People's Republic of China
| | - Defu Zhi
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, 18 Liaohe West Road, Dalian 116600, Liaoning Province People's Republic of China
| | - Shaohui Cui
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, 18 Liaohe West Road, Dalian 116600, Liaoning Province People's Republic of China
| | - Enxia Zhang
- College of Pharmacy, Dalian Medical University, 9 West Section Lvshun South Road, Dalian 116044, Liaoning Province People's Republic of China
| | - Haoming Lan
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, 18 Liaohe West Road, Dalian 116600, Liaoning Province People's Republic of China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Ganjingzi District, Dalian 116024, Liaoning Province People's Republic of China
| | - Zhen Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Ganjingzi District, Dalian 116024, Liaoning Province People's Republic of China
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, 18 Liaohe West Road, Dalian 116600, Liaoning Province People's Republic of China
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, 9 West Section Lvshun South Road, Dalian 116044, Liaoning Province People's Republic of China
| |
Collapse
|
29
|
Kumar K, Kumar Shyamlal BR, Verma R, Kondaiah P, Chaudhary S. Reduction-Triggered Doxorubicin Delivery by Self-Assembled Nanospheres of Lipoylated Caffeine. ChemMedChem 2020; 15:733-737. [PMID: 32162419 DOI: 10.1002/cmdc.202000070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/06/2020] [Indexed: 01/09/2023]
Abstract
This study reports a new amphiphilic bioconjugate (CAFF-LA) derived from the lipoylation of a hydroxyethyl derivative of caffeine. In water, CAFF-LA self-assembles into nanospheres with an average size of 155 nm, as evidenced from dynamic light scattering and electron microscopy studies. The nanospheres are stable in serum and could be disintegrated upon exposure to the reducing environment of dithiothreitol (DTT; 10 mM) and glutathione (GSH; 10 mM). These nanospheres easily encapsulate the chemotherapy medication, doxorubicin (DOX), and demonstrate an efficacious transport into doxorubicin-resistant cervical cancer (HeLa) cells, wherein a marked induction in apoptosis and significantly lower IC50 have been observed when compared to that of free drug. The in vitro assessment of cell viability and hemocompatibility present these nanospheres as potentially safe and efficient intracellular reduction stimulus-responsive drug-delivery vehicles.
Collapse
Affiliation(s)
- Krishan Kumar
- Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur, 302017, India
| | - Bharti Rajesh Kumar Shyamlal
- Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur, 302017, India
| | - Rajbala Verma
- Department of Zoology, University of Rajasthan, Jaipur, 302004, India
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Sandeep Chaudhary
- Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur, 302017, India
| |
Collapse
|
30
|
Wang J, Li D, Fan Y, Shi M, Yang Y, Wang L, Peng Y, Shen M, Shi X. Core-shell tecto dendrimers formed via host-guest supramolecular assembly as pH-responsive intelligent carriers for enhanced anticancer drug delivery. NANOSCALE 2019; 11:22343-22350. [PMID: 31728477 DOI: 10.1039/c9nr08309j] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The design of pH-sensitive supramolecular drug delivery systems for efficient antineoplastic drug delivery remains a huge challenge. Herein, we describe the development of pH-responsive core-shell tecto dendrimers (CSTDs) formed using benzimidazole (BM)-modified generation 3 (G3) poly(amidoamine) (PAMAM) dendrimers (G3.NHAc-BM) as a shell and β-cyclodextrin (CD)-modified G5 PAMAM dendrimers (G5.NHAc-CD) as a core. By virtue of the host-guest recognition and pH-responsiveness of BM/β-CD assembly, the pH-sensitive supramolecular CSTDs of G5.NHAc-CD/BM-G3.NHAc were formed and adopted to encapsulate the anticancer drug doxorubicin (DOX) via hydrophobic interactions for pH-responsive drug delivery applications. The synthesis of dendrimer derivatives and the loading of the DOX were well characterized via different methods. We show that the encapsulated DOX can be released in a sustained manner with a rapid release speed under a slightly acidic pH condition (pH < 6), which is similar to acidic tumor microenvironment. The enhanced intracellular release of DOX and improved anticancer activity of the drug-loaded pH-responsive CSTDs were demonstrated and compared with the control CSTDs formed without pH-responsiveness through flow cytometry and viability assays of cancer cells. Furthermore, the pH-sensitive CSTDs also showed efficient drug penetration and growth inhibition of three-dimensional tumor spheroids owing to the faster DOX release in an acidic pH environment. The pH-sensitive G5.NHAc-CD/BM-G3.NHAc CSTDs may be employed as a valuable intelligent delivery system for various anticancer drugs.
Collapse
Affiliation(s)
- Jianhong Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People's Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Laskar P, Somani S, Campbell SJ, Mullin M, Keating P, Tate RJ, Irving C, Leung HY, Dufès C. Camptothecin-based dendrimersomes for gene delivery and redox-responsive drug delivery to cancer cells. NANOSCALE 2019; 11:20058-20071. [PMID: 31612185 DOI: 10.1039/c9nr07254c] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Combination therapy involving chemotherapeutic drugs and genes is emerging as a promising strategy to provide a synergistic therapeutic effect, to overcome drug resistance while reducing the severe side effects associated with conventional chemotherapeutic drugs. However, the lack of nanomedicines able to simultaneously carry anti-cancer drugs and nucleic acids limits the application of this therapeutic strategy. To overcome this issue, we proposed to synthesize a pro-drug dendrimer by conjugating the PEGylated, positively charged generation 3-diaminobutyric polypropylenimine dendrimer to the anti-cancer drug camptothecin with a redox-sensitive disulphide linkage, and evaluate its efficacy to co-deliver the complexed DNA and camptothecin to cancer cells. This PEGylated pro-drug dendrimer was found to spontaneously self-assemble into cationic (∼3-5 mV) vesicles at pH 7.4, at a critical aggregation concentration of about 200 μg mL-1. These vesicles (dendrimersomes) became smaller (150-200 nm) with increasing dendrimer concentration and remained stable over 7 days. They were able to release about 70% of the conjugated camptothecin in presence of 50 mM glutathione (equivalent to the intracellular environment of tumor tissue). They could also condense more than 85% of the DNA at dendrimer : DNA weight ratios of 5 : 1 and higher. DNA condensation occurred instantly and was found to be stable for at least 24 h. This led to an enhanced cellular uptake of DNA (by up to 1.6-fold) and increased gene transfection (by up to 2.4-fold) in prostate cancer cells in comparison with the unmodified dendrimer. These novel dendrimersomes are therefore promising for single carrier-based combination cancer therapy.
Collapse
Affiliation(s)
- Partha Laskar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Sukrut Somani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Sara Jane Campbell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Margaret Mullin
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Patricia Keating
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Rothwelle J Tate
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Craig Irving
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK
| | - Hing Y Leung
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
32
|
Solubilization of phloretin via steviol glycoside-based solid dispersion and micelles. Food Chem 2019; 308:125569. [PMID: 31644967 DOI: 10.1016/j.foodchem.2019.125569] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 11/22/2022]
Abstract
In this study, the solubility of phloretin (PT) was enhanced via steviol glycoside (STE)-based micelle (MC) and solid dispersion (SD). Computer simulation, characterization, interaction with serum albumin (SA) and in vitro release were carried out to investigate the solubilization mechanisms and the difference in their solubilization capacities. For PT-loaded MC (STE-PT MC), PT was encapsulated into the hydrophobic core of a spherical micelle with a droplet diameter of 5 nm. For PT-loaded SD (STE-PT SD), PT was completely dispersed with the amorphous state in STE. Most of those PTs were directly dissolved in water, and few were encapsulated by STE micelles. The amorphous state combined with relatively large micelles contributed to the high solubilization capacity of STE-PT SD. In addition, PT of STE-PT SD exhibited a higher dissolution rate and more effective interaction with SA than that of STE-PT MC. No undesirable chemical interaction between PT and STE occurred.
Collapse
|
33
|
Tariq I, Pinnapireddy SR, Duse L, Ali MY, Ali S, Amin MU, Goergen N, Jedelská J, Schäfer J, Bakowsky U. Lipodendriplexes: A promising nanocarrier for enhanced gene delivery with minimal cytotoxicity. Eur J Pharm Biopharm 2019; 135:72-82. [DOI: 10.1016/j.ejpb.2018.12.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 12/21/2018] [Accepted: 12/23/2018] [Indexed: 12/11/2022]
|