1
|
Dabas R, Navaratnam N, Iino H, Saidjalolov S, Matile S, Carling D, Rueda DS, Kamaly N. Precise intracellular uptake and endosomal release of diverse functional mRNA payloads via glutathione-responsive nanogels. Mater Today Bio 2025; 30:101425. [PMID: 39839495 PMCID: PMC11745970 DOI: 10.1016/j.mtbio.2024.101425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/23/2025] Open
Abstract
We present a novel, highly customizable glutathione-responsive nanogel (NG) platform for efficient mRNA delivery with precise mRNA payload release control. Optimization of various cationic monomers, including newly synthesized cationic polyarginine, polyhistidine, and acrylated guanidine monomers, allowed fine-tuning of NG properties for mRNA binding. By incorporating a poly(ethylene) glycol-based disulphide crosslinker, we achieved glutathione-triggered mRNA release, enabling targeted intracellular delivery. Our NGs demonstrated superior encapsulation (up to 89.3 %) and loading (10.7 %) efficiencies, with controlled mRNA release kinetics at intracellular glutathione concentrations. NGs outperformed commercial transfection reagents across multiple cell lines, including traditionally difficult-to-transfect lines. We demonstrate the platform's versatility by successfully delivering GFP mRNA, Mango II RNA aptamers, and functionally relevant β2-AMPK mRNA. Furthermore, we used TIRF microscopy to measure exact RNA copy number within the NGs. Notably, mechanistic cellular uptake studies revealed that disulphide-containing NGs exhibit enhanced cellular uptake and endosomal escape, potentially due to interactions with cell surface thiols. This work represents a highly tuneable, efficient, and biocompatible platform for mRNA delivery with relevance for gene therapy and vaccine development.
Collapse
Affiliation(s)
- Rupali Dabas
- Cellular Stress Research Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, W12 0BZ, London, UK
| | - Naveenan Navaratnam
- Cellular Stress Research Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
| | - Haruki Iino
- Single Molecule Imaging Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
- Section of Virology, Department of Infectious Disease, Imperial College London, W12 0HS, London, UK
| | | | - Stefan Matile
- Department of Organic Chemistry, University of Geneva, Geneva, Switzerland
| | - David Carling
- Cellular Stress Research Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
| | - David S. Rueda
- Single Molecule Imaging Group, MRC Laboratory of Medical Sciences, Imperial College London, W12 0HS, London, UK
- Section of Virology, Department of Infectious Disease, Imperial College London, W12 0HS, London, UK
| | - Nazila Kamaly
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, W12 0BZ, London, UK
| |
Collapse
|
2
|
Musa M, Sun X, Shi J, Li J, Zhang S, Shi X. Intelligent responsive nanogels: New Horizons in cancer therapy. Int J Pharm 2025; 669:125050. [PMID: 39645062 DOI: 10.1016/j.ijpharm.2024.125050] [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: 08/24/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Biologically engineered nanogels formed through sophisticated intramolecular crosslinking processes represent the forefront of next-generation drug delivery systems. These innovative systems offer many advantages, like adjustable size, satisfactory biocompatibility, and minimal toxicity. Their unique attributes facilitate deep penetration and long-term retention of drugs in tumors, effectively enhancing the anti-tumor effects. Nonetheless, the rapid disintegration of nanogels and the subsequent triggering of drug release at the tumor site pose significant challenges in achieving more effective and precise tumor treatments. Therefore, increasing research has been dedicated to exploring stimulus-responsive nanogels for enhancing tumor therapy. This review aims to encapsulate the research advancements in emerging stimulus-responsive antitumor nanogels. Firstly, a detailed exposition is provided on various endogenous stimulus-responsive nanogels, encompassing factors such as pH, hypoxia, enzymes, reactive oxygen species (ROS), and glutathione (GSH). Secondly, various nanogels triggered by exogenous stimuli such as light, ultrasound, temperature, and magnetic fields are elaborately presented. Furthermore, nanogels with multifaceted stimulus-responsive properties are also skillfully designed. Finally, the future directions, application prospects, and challenges of intelligent responsive nanogels in cancer treatment are highlighted.
Collapse
Affiliation(s)
- MiriGuli Musa
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xinxin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jianbin Shi
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jing Li
- School of Pharmacy, Shenyang Medical College, Shenyang, Liaoning Province, China.
| | - Shenwu Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
3
|
Ciftci F, Özarslan AC, Kantarci İC, Yelkenci A, Tavukcuoglu O, Ghorbanpour M. Advances in Drug Targeting, Drug Delivery, and Nanotechnology Applications: Therapeutic Significance in Cancer Treatment. Pharmaceutics 2025; 17:121. [PMID: 39861768 PMCID: PMC11769154 DOI: 10.3390/pharmaceutics17010121] [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: 12/03/2024] [Revised: 01/01/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
In the 21st century, thanks to advances in biotechnology and developing pharmaceutical technology, significant progress is being made in effective drug design. Drug targeting aims to ensure that the drug acts only in the pathological area; it is defined as the ability to accumulate selectively and quantitatively in the target tissue or organ, regardless of the chemical structure of the active drug substance and the method of administration. With drug targeting, conventional, biotechnological and gene-derived drugs target the body's organs, tissues, and cells that can be selectively transported to specific regions. These systems serve as drug carriers and regulate the timing of release. Despite having many advantageous features, these systems have limitations in thoroughly treating complex diseases such as cancer. Therefore, combining these systems with nanoparticle technologies is imperative to treat cancer at both local and systemic levels effectively. The nanocarrier-based drug delivery method involves encapsulating target-specific drug molecules into polymeric or vesicular systems. Various drug delivery systems (DDS) were investigated and discussed in this review article. The first part discussed active and passive delivery systems, hydrogels, thermoplastics, microdevices and transdermal-based drug delivery systems. The second part discussed drug carrier systems in nanobiotechnology (carbon nanotubes, nanoparticles, coated, pegylated, solid lipid nanoparticles and smart polymeric nanogels). In the third part, drug targeting advantages were discussed, and finally, market research of commercial drugs used in cancer nanotechnological approaches was included.
Collapse
Affiliation(s)
- Fatih Ciftci
- Department of Biomedical Engineering, Faculty of Engineering, Fatih Sultan Mehmet Vakıf University, Istanbul 34015, Turkey
- Department of Technology Transfer Office, Fatih Sultan Mehmet Vakıf University, Istanbul 34015, Turkey
| | - Ali Can Özarslan
- Department of Metallurgical and Materials Engineering, Istanbul University-Cerrahpasa, Istanbul 34320, Turkey;
| | - İmran Cagri Kantarci
- Department of Bioengineering, Faculty of Chemistry-Metallurgy, Yildiz Technical University, Istanbul 34210, Turkey;
| | - Aslihan Yelkenci
- Department of Pediatric Dentistry, Faculty of Dentistry, University of Health Sciences, Istanbul 34668, Turkey;
| | - Ozlem Tavukcuoglu
- Department of Biochemistry, Faculty of Hamidiye Pharmacy, University of Health Sciences, Istanbul 34668, Turkey;
| | - Mansour Ghorbanpour
- Department of Medicinal Plants, Faculty of Agriculture and Natural Resources, Arak University, Arak 38156-8-8349, Iran;
| |
Collapse
|
4
|
Sojdeh S, Safarkhani M, Daneshgar H, Aldhaher A, Heidari G, Nazarzadeh Zare E, Iravani S, Zarrabi A, Rabiee N. Promising breakthroughs in amyotrophic lateral sclerosis treatment through nanotechnology's unexplored frontier. Eur J Med Chem 2025; 282:117080. [PMID: 39577228 DOI: 10.1016/j.ejmech.2024.117080] [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: 08/24/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/24/2024]
Abstract
This review explores the transformative potential of nanotechnology in the treatment and diagnosis of amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disorder characterized by motor neuron degeneration, muscle weakness, and eventual paralysis. Nanotechnology offers innovative solutions across various domains, including targeted drug delivery, neuroprotection, gene therapy and editing, biomarker detection, advanced imaging techniques, and tissue engineering. By enhancing the precision and efficacy of therapeutic interventions, nanotechnology facilitates key advancements such as crossing the blood-brain barrier, targeting specific cell types, achieving sustained therapeutic release, and enabling combination therapies tailored to the complex pathophysiology of ALS. Despite its immense promise, the clinical translation of these approaches faces challenges, including potential cytotoxicity, biocompatibility, and regulatory compliance, which must be addressed through rigorous research and testing. This review emphasizes the application of nanotechnology in targeted drug delivery and gene therapy/editing for ALS, drawing on the author's prior work with various nanotechnological platforms to illustrate strategies for overcoming similar obstacles in drug and gene delivery. By bridging the gap between cutting-edge technology and clinical application, this article aims to highlight the vital role of nanotechnology in shaping the future of ALS treatment.
Collapse
Affiliation(s)
- Soheil Sojdeh
- Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Moein Safarkhani
- Department of Biological Sciences and Bioengineering, Nano-Bio High-Tech Materials Research Center, Inha University, Incheon, 22212, Republic of Korea
| | - Hossein Daneshgar
- Department of Inorganic Chemistry, Faculty of Chemistry and Petroleum Sciences, Shahid Beheshti University, P. O. Box 19839-63113, Tehran, Iran
| | - Abdullah Aldhaher
- Department of Chemistry, Sharif University of Technology, Tehran, Iran
| | - Golnaz Heidari
- School of Natural Sciences, Massey University, Private Bag 11 222, Palmerston North, 4410, New Zealand
| | - Ehsan Nazarzadeh Zare
- School of Chemistry, Damghan University, Damghan, 36716-45667, Iran; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | | | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396, Istanbul, Turkey; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077, India.
| |
Collapse
|
5
|
Guo J, Qiu Y, Zhang J, Xue C, Zhu J. A review on polysaccharide-based delivery systems for edible bioactives: pH responsive, controlled release, and emerging applications. Int J Biol Macromol 2024; 291:139178. [PMID: 39730044 DOI: 10.1016/j.ijbiomac.2024.139178] [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: 09/01/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
pH changes occur during bodily lesions, presenting an opportunity for leveraging pH-responsive delivery systems as signals for a targeted response. This review explores the design and application of pH-responsive delivery systems based on natural polysaccharides for the controlled release of bioactives. The article examines the development of diverse delivery carriers, including nanoparticles, nanofibers, nanogels, core-shell carriers, hydrogels, emulsions as well as liposomes and their capacity to respond to pH variations, enabling the precise and targeted delivery of bioactives within the human body. These polysaccharide-based delivery systems can be made pH-responsive by modulating the charge of polybasic or polyacidic polysaccharides, inducing swelling of the carrier and subsequent release of the encapsulated bioactives. These pH-responsive systems show promise in stabilizing under acidic conditions for enhanced retention in the stomach during oral delivery while also enabling targeted release at low pH sites such as tumors and wounds, thereby accelerating wound healing and aiding in cancer therapy and inflammation treatment. pH can co-respond with a variety of stimuli, including temperature, enzymes and reactive oxygen species, enabling more precise responses to the microenvironment for targeted delivery. It provides solid theoretical foundations for the advancement of personalized nutrition and therapeutics through controlled and responsive release technologies.
Collapse
Affiliation(s)
- Jiaxin Guo
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China
| | - Yang Qiu
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhang
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China
| | - Chenxu Xue
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China
| | - Jiajin Zhu
- College of Biosystems Engineering & Food Science, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
6
|
Khalaf MM, Gouda M, Abou Taleb MF, Heakal FET, Abd El-Lateef HM. Fabrication of smart nanogel based on carrageenan and green coffee extract as a long-term antifouling agent to improve biofilm prevention in food production. Food Chem 2024; 461:140719. [PMID: 39146677 DOI: 10.1016/j.foodchem.2024.140719] [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: 03/27/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024]
Abstract
This study investigates the extract of the bioactive compounds from green coffee extract (GCE) and the loading of two different concentrations of GCE (1% and 2%) onto carrageenan nanogels (CAR NGs) to compare their antibacterial and antibiofilm effects with unloaded nanogels (NGs). The bioactive compounds of GCE were characterized using GC-MS analysis. The GCE1 and GCE2 were successfully deposited onto the surface of CAR NGs. The antibacterial and antibiofilm potential of prepared NGs were conducted against some foodborne pathogens (E. coli O157, Salmonella enterica, Staphylococcus aureus, and Listeria monocytogenes). The results of GC-MS analysis indicated that there were identified 16 bioactive compounds in GCE, including caffeine (36.27%), Dodemorph (9.04%), and D-Glycero-d-ido-heptose (2.44%), contributing to its antimicrobial properties. The antibacterial coatings demonstrated a notable antimicrobial effect, showing zone of inhibition (ZOI) diameters of up to 37 mm for GCE2 loaded CAR NGs. The minimum inhibitory concentration (MIC) values for GCE2 loaded CAR NGs were 80 ppm for E. coli O157, and 120 ppm for S. enterica, S. aureus, and L. monocytogenes, achieving complete bacterial inactivation within 10-15 min of exposure. Both GCE1 and GCE2 loaded CAR NGs significantly reduced biofilm cell densities on stainless steel (SS) materials for E. coli O157, S. enterica, S. aureus, and L. monocytogenes, with reductions ranging from 60% to 95%. Specifically, biofilm densities were reduced by up to 95% for E. coli O157, 89% for S. enterica, 85% for S. aureus, and 80% for L. monocytogenes. Results of the toxicity evaluation indicated that the NGs were non-toxic and biocompatible, with predicted EC50 values proved their biocompatibility and safety. These results recommended that GCE loaded CAR NGs are promising as natural antimicrobial agents for enhancing food safety and extending shelf life. Further, the study concluded that incorporating GCE into CAR NGs is an effective strategy for developing sustainable antimicrobial coatings for the food industry and manufacturing.
Collapse
Affiliation(s)
- Mai M Khalaf
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia; Department of Chemistry, Faculty of Science, Sohag University, Sohag 82524, Egypt.
| | - Mohamed Gouda
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
| | - Manal F Abou Taleb
- Department of Chemistry, College of Science and Humanities, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Hany M Abd El-Lateef
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia; Department of Chemistry, Faculty of Science, Sohag University, Sohag 82524, Egypt
| |
Collapse
|
7
|
Cai J, Zhong H, Luo J, Huang X, Xu Q, Li P. Inhalable multi-stimulus sensitive curcumin-alginate nanogels for scavenging reactive oxygen species and anti-inflammatory co-ordination to alleviate acute lung injury. Int J Biol Macromol 2024; 283:137816. [PMID: 39571867 DOI: 10.1016/j.ijbiomac.2024.137816] [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: 08/26/2024] [Revised: 11/08/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024]
Abstract
Acute lung injury (ALI) is one of the most common and extremely critical clinical conditions, which progresses with an inflammatory response and overproduction of reactive oxygen species (ROS), leading to oxidative damage to the lungs. Curcumin (Cur) has great potential in treating ALI due to its excellent antioxidant and anti-inflammatory effects. In this study, Cur and alginate were cross-linked by zinc ions and intermolecular hydrogen bonding to form an inhalable aqueous nanogel system to overcome Cur's low solubility and bioavailability. Cur-alginate (ZA-Cur) nanogels exhibited superior antioxidant properties and down-regulated inflammation-associated factors in vitro with controlled-release behavior under multi-stimulus conditions such as temperature, pH, and ions. Meanwhile, the nanogels system could effectively scavenge cellular ROS to repair oxidative stress damage. In a mice model of ALI, tracheal nebulised inhalation of ZA-Cur nanogels down-regulated the expression of inflammation-related genes such as TNF-α, IL-1β, and IL-6, as well as modulated MDA content and CAT activity to attenuate oxidative stress injury, showing promising lung-protective effects. In conclusion, this work developed inhalable ZA-Cur nanogels to decelerate the progression of lesions in ALI by scavenging intracellular ROS and alleviating inflammation simultaneously, which may be a promising strategy for treating ALI.
Collapse
Affiliation(s)
- Jinyun Cai
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Haiyi Zhong
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China.
| | - Jianwei Luo
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Xinghai Huang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Qiuting Xu
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Peiyuan Li
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China.
| |
Collapse
|
8
|
Vashist A, Perez Alvarez G, Andion Camargo V, Raymond AD, Arias AY, Kolishetti N, Vashist A, Manickam P, Aggarwal S, Nair M. Recent advances in nanogels for drug delivery and biomedical applications. Biomater Sci 2024; 12:6006-6018. [PMID: 39484856 PMCID: PMC11528912 DOI: 10.1039/d4bm00224e] [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: 02/12/2024] [Accepted: 06/26/2024] [Indexed: 11/03/2024]
Abstract
Nanotechnology has shown great promise for researchers to develop efficient nanocarriers for better therapy, imaging, and sustained release of drugs. The existing treatments are accompanied by serious toxicity limitations, leading to severe side effects, multiple drug resistance, and off-target activity. In this regard, nanogels have garnered significant attention for their multi-functional role combining advanced therapeutics with imaging in a single platform. Nanogels can be functionalized to target specific tissues which can improve the efficiency of drug delivery and other challenges associated with the existing nanocarriers. Translation of nanogel technology requires more exploration towards stability and enhanced efficiency. In this review, we present the advances and challenges related to nanogels for cancer therapy, ophthalmology, neurological disorders, tuberculosis, wound healing, and anti-viral applications. A perspective on recent research trends of nanogels for translation to clinics is also discussed.
Collapse
Affiliation(s)
- Arti Vashist
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
| | - Gabriela Perez Alvarez
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| | - Vianessa Andion Camargo
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| | - Andrea D Raymond
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| | - Adriana Yndart Arias
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| | - Nagesh Kolishetti
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| | - Atul Vashist
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, India
- Centre of Excellence in Nanosensors and Nanomedicine, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh, India
| | - Pandiaraj Manickam
- Electrodics and Electrocatalysis Division, CSIR-Central Electrochemical Research Institute (CECRI), Karaikudi 630 003, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Saurabh Aggarwal
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| | - Madhavan Nair
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
9
|
Huang Z, Dong H, Qiu Y, Chang A, Zhu H. pH-Responsive Deacetylated Sphingan WL Gum-Based Microgels for the Oral Delivery of Ciprofloxacin Hydrochloride. ACS OMEGA 2024; 9:46397-46407. [PMID: 39583669 PMCID: PMC11579934 DOI: 10.1021/acsomega.4c07589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024]
Abstract
Sphingan WL gum (WL) is an extracellular polysaccharide with a carboxyl group produced by Sphingomonas sp. WG. Recently, we have successfully obtained deacetylated WL (DWL) with good water solubility by alkaline treatment. In this study, a DWL-based microgel (named DWLM) with semi-interpenetrating network structure was constructed for the first time and used to deliver the oral drug ciprofloxacin hydrochloride (CIP). DLS results suggested that DWLM had a dual response to pH and temperature. The in vitro cumulative drug release curves showed that the amount of CIP released from the microgel was higher at pH 6.8 than that at pH 3.0. Biocompatibility assessments using HEK293T showed that cell viability was 75.9 ± 1.7% at the DWLM-CIP concentration of 4 mg/mL. While, the cell viability of CIP at the same concentration was only 54.9 ± 1.0%, indicating that DWLM-CIP has good biocompatibility. Antimicrobial performance tests revealed that DWLM-CIP at a concentration of 1 mg/mL could effectively inhibit the growth of Escherichia coli for up to 4 days. When the concentration of DWLM-CIP reached 4 mg/mL, the growth of Staphylococcus aureus was effectively suppressed for up to 3 days, demonstrating the long-lasting antimicrobial efficacy of DWLM-CIP. All of these results indicate that DWL-based microgels have great potential as oral drug delivery carriers.
Collapse
Affiliation(s)
- Zhenyin Huang
- Fujian-Taiwan Science and
Technology Cooperation Base of Biomedical Materials and Tissue Engineering,
Engineering Research Center of Industrial Biocatalysis, Fujian Provincial
Key Laboratory of Advanced Materials Oriented Chemical Engineering,
Fujian Provincial Key Laboratory of Polymer Materials, College of
Chemistry and Materials Science, Fujian
Normal University, Fuzhou 350117, People’s
Republic of China
| | - Hanyu Dong
- Fujian-Taiwan Science and
Technology Cooperation Base of Biomedical Materials and Tissue Engineering,
Engineering Research Center of Industrial Biocatalysis, Fujian Provincial
Key Laboratory of Advanced Materials Oriented Chemical Engineering,
Fujian Provincial Key Laboratory of Polymer Materials, College of
Chemistry and Materials Science, Fujian
Normal University, Fuzhou 350117, People’s
Republic of China
| | - Yingjie Qiu
- Fujian-Taiwan Science and
Technology Cooperation Base of Biomedical Materials and Tissue Engineering,
Engineering Research Center of Industrial Biocatalysis, Fujian Provincial
Key Laboratory of Advanced Materials Oriented Chemical Engineering,
Fujian Provincial Key Laboratory of Polymer Materials, College of
Chemistry and Materials Science, Fujian
Normal University, Fuzhou 350117, People’s
Republic of China
| | - Aiping Chang
- Fujian-Taiwan Science and
Technology Cooperation Base of Biomedical Materials and Tissue Engineering,
Engineering Research Center of Industrial Biocatalysis, Fujian Provincial
Key Laboratory of Advanced Materials Oriented Chemical Engineering,
Fujian Provincial Key Laboratory of Polymer Materials, College of
Chemistry and Materials Science, Fujian
Normal University, Fuzhou 350117, People’s
Republic of China
| | - Hu Zhu
- Fujian-Taiwan Science and
Technology Cooperation Base of Biomedical Materials and Tissue Engineering,
Engineering Research Center of Industrial Biocatalysis, Fujian Provincial
Key Laboratory of Advanced Materials Oriented Chemical Engineering,
Fujian Provincial Key Laboratory of Polymer Materials, College of
Chemistry and Materials Science, Fujian
Normal University, Fuzhou 350117, People’s
Republic of China
| |
Collapse
|
10
|
Hajebi S, Chamanara M, Nasiri SS, Ghasri M, Mouraki A, Heidari R, Nourmohammadi A. Advances in stimuli-responsive gold nanorods for drug-delivery and targeted therapy systems. Biomed Pharmacother 2024; 180:117493. [PMID: 39353321 DOI: 10.1016/j.biopha.2024.117493] [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: 06/15/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
In recent years, the use of gold nanorods (AuNRs) has garnered considerable attention in biomedical applications due to their unique optical and physicochemical properties. They have been considered as potential tools for the advanced treatment of diseases by various stimuli such as magnetic fields, pH, temperature and light in the fields of targeted therapy, imaging and drug delivery. Their biocompatibility and tunable plasmonic properties make them a versatile platform for a range of biomedical applications. While endogenous stimuli have limited cargo delivery control at specific sites, exogenous stimuli are a more favored approach despite their circumscribed penetration depth for releasing the cargo at the specific target. Dual/multi-stimuli responsive AuNTs can be triggered by multiple stimuli for enhanced control and specificity in biomedical applications. This review provides to provide a summary of the biomedical applications of stimuli-responsive AuNRs, including their endogenous and exogenous properties, as well as their dual/multi-functionality and potential for clinical delivery. This review provides a comprehensive review on the improvement of therapeutic efficacy and the effective control of drug release with AuNRs, highlights AuNRs design strategies in recent years, discusses the advantages or challenges so far in the field of AuNRs. Finally, we have addressed the clinical translation bio-integrated nanoassemblies (CTBNs) in this field.
Collapse
Affiliation(s)
- Sakineh Hajebi
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran; Biomaterial and Medicinal Chemistry Research Center, AJA University of Medical Science, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran; Biomaterial and Medicinal Chemistry Research Center, AJA University of Medical Science, Tehran, Iran
| | - Shadi Sadat Nasiri
- Department of Polymer Engineering and Color Technology, Amirkabir University of Technology, Tehran, Iran
| | - Mahsa Ghasri
- Adhesive and Resin Department, Polymer Processing Faculty, Iran Polymer and Petrochemical Institute (IPPI), Tehran, Iran
| | - Alireza Mouraki
- Department of Surface Coating and Corrosion, Institute for Color Science and Technology, Tehran, Iran
| | - Reza Heidari
- Cancer Epidemiology Research Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran; Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran; Biomaterial and Medicinal Chemistry Research Center, AJA University of Medical Science, Tehran, Iran.
| | - Abbas Nourmohammadi
- Clinical Biomechanics and Ergonomics Research Center, AJA University of Medical Sciences, Tehran, Iran; Research Center of Aerospace Medicine, AJA University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Lv J, Chen J, Song Y, Yao Y, Wu G, Yuan D, Gu X, Li X, Xu C, Zhou B, Ye M, Lv T, Wang D, Song Y. Co-Delivery of VEGF siRNA and THPP via Metal-Organic Framework Reverses Cisplatin-Resistant Non-Small Cell Lung Cancer and Inhibits Metastasis through a MUC4 Regulating Mechanism. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56910-56925. [PMID: 39397733 DOI: 10.1021/acsami.4c15175] [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: 10/15/2024]
Abstract
Cisplatin resistance significantly impacts the antitumor efficacy of cisplatin chemotherapy and contributes to poor prognosis, including metastasis. In this study, we present the utilization of metal-organic framework (MOF) nanoparticles as the therapeutic component and drug loading scaffold for implementing a ternary combination therapeutic strategy to combat cisplatin-resistant lung cancer and metastasis. Specifically, by engineering MOFs (Cis@MOF-siVEGF) through the self-assembly of THPP as photosensitizer for photodynamic therapy (PDT), along with the incorporation of cisplatin (DDP) and VEGF siRNA (siVEGF), we propose the leverage of photodynamic-induced oxidative damage and gene silencing of the angiogenic factor to reverse cisplatin resistance and sensitize therapeutic potency. Our findings demonstrated that the chemo/photodynamic/antiangiogenic triple combination therapy via Cis@MOF-siVEGF under irradiation effectively inhibits cisplatin-resistant tumor growth and induces abscopal effects. Importantly, molecular mechanistic exploration suggested that MUC4 exerted regulatory effects on governing cancer metastasis, thus representing a potential immunotherapeutic target for cancer intervention. Overall, our study creates a MOFs-based multicomponent delivery platform for complementary therapeutic modules with synergistically enhanced antitumor efficacy and sheds light on potential regulatory mechanisms on cisplatin-resistance cancers.
Collapse
Affiliation(s)
- Jiawen Lv
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Jiayan Chen
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Yueyue Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing 210002, People's Republic of China
| | - Yanwen Yao
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Guannan Wu
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Dongmei Yuan
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Xiaoling Gu
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Xing Li
- Department of Endocrinology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Chunwei Xu
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, People's Republic of China
| | - Baolong Zhou
- School of Pharmacy, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing 210002, People's Republic of China
| | - Dong Wang
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing 210002, People's Republic of China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University of Chinese Medicine, Nanjing 210002, People's Republic of China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing 210002, People's Republic of China
| |
Collapse
|
12
|
Zhao Y, Cao Z, Zhang J, Tian J, Cai H. Thermo-responsible PNIPAM-grafted polystyrene microspheres for mesenchymal stem cells culture and detachment. Biomed Mater 2024; 19:065023. [PMID: 39312938 DOI: 10.1088/1748-605x/ad7e6e] [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: 07/10/2024] [Accepted: 09/23/2024] [Indexed: 09/25/2024]
Abstract
The preparation of cells is a critical step in cell therapy. To ensure the effectiveness of cells used for clinical treatments, it is essential to harvest adherent cells from the culture media in a way that preserves their high viability and full functionality. In this study, we developed temperature-responsive poly(N-isopropylacrylamide) (PNIPAM)-grafted polystyrene (PS) microspheres using reversible addition-fragmentation chain transfer polymerization. These microspheres allow for the non-destructive harvesting of cultured cells through temperature changes. The composition and physicochemical properties of the PNIPAM-grafted PS microspheres were confirmed using infrared spectroscopy, elemental analysis, dynamic light scattering, and thermogravimetric analysis.In vitroexperiments demonstrated that these microspheres exhibit excellent biocompatibility, supporting the adhesion and proliferation of various cells. Moreover, the microspheres showed good temperature responsiveness in thermosensitive detachment experiments with GFP-HepG2cells and umbilical cord mesenchymal stem cells (UC-MSCs). Additionally, through orthogonal experiments, we identified a cell detachment aid mixture that significantly improved the dispersibility of cells detached from the microspheres, enhancing the efficiency of thermosensitive cell detachment by approximately 40%. The harvested UC-MSCs retained their capacity for re-proliferation and trilineage differentiation. Consequently, the temperature-responsive microspheres developed in this study, combined with the cell detachment aid mixtures, hold great potential for large-scale culture and harvesting of therapeutic cells in clinical applications.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Zida Cao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Jingwei Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Jia Tian
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| | - Haibo Cai
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, People's Republic of China
| |
Collapse
|
13
|
Hu Y, Zhou Y, Li K, Zhou D. Recent advances in near-infrared stimulated nanohybrid hydrogels for cancer photothermal therapy. Biomater Sci 2024; 12:4590-4606. [PMID: 39136645 DOI: 10.1039/d4bm00662c] [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: 09/11/2024]
Abstract
Nanomedicine has emerged as a promising avenue for advancing cancer treatment, but the challenge of mitigating its in vivo side effects necessitates the development of innovative structures and materials. Recent investigation has unveiled nanogels as particularly compelling candidates, characterized by a porous, three-dimensional network architecture that exhibits exceptional drug loading capacity. Beyond this, nanogels boast a substantial specific surface area and can be tailored with specific chemical functionalities. Consequently, nanogels are frequently engineered as a multi-modal synergistic platform for combating cancer, wherein photothermal therapy stands out due to its capacity to penetrate deep tissues and achieve localized tumor eradication through the application of elevated temperatures. In this review, we delve into the synthesis of diverse varieties of photothermal nanogels capable of controlled drug release triggered by either chemical or physical stimuli. It also summarizes their potential for synergistic integration with photothermal therapy alongside other therapeutic modalities to realize effective tumor ablation. Moreover, we analyze the primary mechanisms underlying the contribution of photothermal nanogels to cancer treatment while underscoring their adeptness in regulating therapeutic temperatures for repairing bone defects resulting from tumor-associated trauma. Envisioned as an auspicious strategy in the realm of cancer therapy, photothermal nanogels hold promise for furnishing controlled drug delivery and precise thermal ablation capabilities.
Collapse
Affiliation(s)
- Yongjun Hu
- Department of Oncology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yi Zhou
- Huanggang Central Hospital of Yangtze University, Huanggang, 438000, China
| | - Kaichun Li
- Department of Oncology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
| | - Dong Zhou
- Engineering Research Centre for Biomedical Materials of Ministry of Education, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
14
|
Lu H, Cai Z, Hu P. Recent Advances in Polymeric Delivery Vehicles for Controlled and Sustained Drug Release. Pharmaceutics 2024; 16:1184. [PMID: 39339220 PMCID: PMC11435192 DOI: 10.3390/pharmaceutics16091184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
In the realm of modern therapeutics, the development of polymeric delivery vehicles has revolutionized drug administration, offering a sophisticated approach to controlled and sustained drug release [...].
Collapse
Affiliation(s)
- Hong Lu
- Department of Burns & Plastic Surgery, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| | - Zheng Cai
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ping Hu
- Department of Burns & Plastic Surgery, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou 510006, China
- College of Pharmacy, Jinan University, Guangzhou 510006, China
| |
Collapse
|
15
|
Ghosh Majumdar A, Pany B, Parua SS, Mukherjee D, Panda A, Mohanty M, Das B, Si S, Mohanty PS. Stimuli-Responsive Nanogel/Microgel Hybrids as Targeted Drug Delivery Systems: A Comprehensive Review. BIONANOSCIENCE 2024; 14:3496-3521. [DOI: 10.1007/s12668-024-01577-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 01/06/2025]
|
16
|
Ji XY, Zou YX, Lei HF, Bi Y, Yang R, Tang JH, Jin QR. Advances in Cyclodextrins and Their Derivatives in Nano-Delivery Systems. Pharmaceutics 2024; 16:1054. [PMID: 39204399 PMCID: PMC11360519 DOI: 10.3390/pharmaceutics16081054] [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: 05/16/2024] [Revised: 07/14/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
The diversity of cyclodextrins and their derivatives is increasing with continuous research. In addition to monomolecular cyclodextrins with different branched chains, cyclodextrin-based polymers have emerged. The aim of this review is to summarize these innovations, with a special focus on the study of applications of cyclodextrins and their derivatives in nano-delivery systems. The areas covered include nanospheres, nano-sponges, nanogels, cyclodextrin metal-organic frameworks, liposomes, and emulsions, providing a comprehensive and in-depth understanding of the design and development of nano-delivery systems.
Collapse
Affiliation(s)
- Xin-Yu Ji
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China;
| | - Yi-Xuan Zou
- National institute of Metrology, Beijing 100029, China
| | - Han-Fang Lei
- College of Pharmacy, Anhui Medical University, Hefei 230032, China; (H.-F.L.); (Y.B.)
| | - Yong Bi
- College of Pharmacy, Anhui Medical University, Hefei 230032, China; (H.-F.L.); (Y.B.)
| | - Rui Yang
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing 100050, China;
| | - Ji-Hui Tang
- College of Pharmacy, Anhui Medical University, Hefei 230032, China; (H.-F.L.); (Y.B.)
| | - Qing-Ri Jin
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310053, China;
| |
Collapse
|
17
|
Desai N, Chavda V, Singh TRR, Thorat ND, Vora LK. Cancer Nanovaccines: Nanomaterials and Clinical Perspectives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401631. [PMID: 38693099 DOI: 10.1002/smll.202401631] [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: 03/01/2024] [Revised: 03/30/2024] [Indexed: 05/03/2024]
Abstract
Cancer nanovaccines represent a promising frontier in cancer immunotherapy, utilizing nanotechnology to augment traditional vaccine efficacy. This review comprehensively examines the current state-of-the-art in cancer nanovaccine development, elucidating innovative strategies and technologies employed in their design. It explores both preclinical and clinical advancements, emphasizing key studies demonstrating their potential to elicit robust anti-tumor immune responses. The study encompasses various facets, including integrating biomaterial-based nanocarriers for antigen delivery, adjuvant selection, and the impact of nanoscale properties on vaccine performance. Detailed insights into the complex interplay between the tumor microenvironment and nanovaccine responses are provided, highlighting challenges and opportunities in optimizing therapeutic outcomes. Additionally, the study presents a thorough analysis of ongoing clinical trials, presenting a snapshot of the current clinical landscape. By curating the latest scientific findings and clinical developments, this study aims to serve as a comprehensive resource for researchers and clinicians engaged in advancing cancer immunotherapy. Integrating nanotechnology into vaccine design holds immense promise for revolutionizing cancer treatment paradigms, and this review provides a timely update on the evolving landscape of cancer nanovaccines.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Telangana, 502285, India
| | - Vivek Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, 380009, India
| | | | - Nanasaheb D Thorat
- Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
- Department of Physics, Bernal Institute, Castletroy, Limerick, V94T9PX, Ireland
- Nuffield Department of Women's & Reproductive Health, Medical Science Division, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| |
Collapse
|
18
|
Mastella P, Todaro B, Luin S. Nanogels: Recent Advances in Synthesis and Biomedical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1300. [PMID: 39120405 PMCID: PMC11314474 DOI: 10.3390/nano14151300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024]
Abstract
In the context of advanced nanomaterials research, nanogels (NGs) have recently gained broad attention for their versatility and promising biomedical applications. To date, a significant number of NGs have been developed to meet the growing demands in various fields of biomedical research. Summarizing preparation methods, physicochemical and biological properties, and recent applications of NGs may be useful to help explore new directions for their development. This article presents a comprehensive overview of the latest NG synthesis methodologies, highlighting advances in formulation with different types of hydrophilic or amphiphilic polymers. It also underlines recent biomedical applications of NGs in drug delivery and imaging, with a short section dedicated to biosafety considerations of these innovative nanomaterials. In conclusion, this article summarizes recent innovations in NG synthesis and their numerous applications, highlighting their considerable potential in the biomedical field.
Collapse
Affiliation(s)
- Pasquale Mastella
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
- Fondazione Pisana per la Scienza ONLUS, Via Ferruccio Giovannini 13, 56017 San Giuliano Terme, PI, Italy
| | - Biagio Todaro
- Department of Chemical Engineering, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium;
| | - Stefano Luin
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy
- NEST Laboratory, Istituto Nanoscienze-CNR, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
19
|
Xie B, Xie H. Application of stimuli-responsive hydrogel in brain disease treatment. Front Bioeng Biotechnol 2024; 12:1450267. [PMID: 39091971 PMCID: PMC11291207 DOI: 10.3389/fbioe.2024.1450267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024] Open
Abstract
Treating brain diseases presents significant challenges due to neuronal degeneration, inflammation, and the intricate nature of the brain. Stimuli-responsive hydrogels, designed to closely resemble the brain's extracellular matrix, have emerged as promising candidates for controlled drug delivery and tissue engineering. These hydrogels have the unique ability to encapsulate therapeutic agents and release them in a controlled manner when triggered by environmental stimuli. This property makes them particularly suitable for delivering drugs precisely to targeted areas of the brain, while minimizing collateral damage to healthy tissue. Their preclinical success in treating various brain diseases in animal studies underscores their translational potential for human brain disease treatment. However, a deeper understanding of their long-term behavior, biodistribution, and biocompatibility within the brain remains crucial. Furthermore, exploring novel hydrogel systems and therapeutic combinations is paramount for advancing towards more effective treatments. This review summarizes the latest advancements in this field over the past 5 years, specifically highlighting preclinical progress with novel stimuli-responsive hydrogels for treating brain diseases.
Collapse
Affiliation(s)
- Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, Sichuan, China
| | - Huangfan Xie
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
20
|
Caturano A, Nilo R, Nilo D, Russo V, Santonastaso E, Galiero R, Rinaldi L, Monda M, Sardu C, Marfella R, Sasso FC. Advances in Nanomedicine for Precision Insulin Delivery. Pharmaceuticals (Basel) 2024; 17:945. [PMID: 39065795 PMCID: PMC11279564 DOI: 10.3390/ph17070945] [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: 06/04/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Diabetes mellitus, which comprises a group of metabolic disorders affecting carbohydrate metabolism, is characterized by improper glucose utilization and excessive production, leading to hyperglycemia. The global prevalence of diabetes is rising, with projections indicating it will affect 783.2 million people by 2045. Insulin treatment is crucial, especially for type 1 diabetes, due to the lack of β-cell function. Intensive insulin therapy, involving multiple daily injections or continuous subcutaneous insulin infusion, has proven effective in reducing microvascular complications but poses a higher risk of severe hypoglycemia. Recent advancements in insulin formulations and delivery methods, such as ultra-rapid-acting analogs and inhaled insulin, offer potential benefits in terms of reducing hypoglycemia and improving glycemic control. However, the traditional subcutaneous injection method has drawbacks, including patient compliance issues and associated complications. Nanomedicine presents innovative solutions to these challenges, offering promising avenues for overcoming current drug limitations, enhancing cellular uptake, and improving pharmacokinetics and pharmacodynamics. Various nanocarriers, including liposomes, chitosan, and PLGA, provide protection against enzymatic degradation, improving drug stability and controlled release. These nanocarriers offer unique advantages, ranging from enhanced bioavailability and sustained release to specific targeting capabilities. While oral insulin delivery is being explored for better patient adherence and cost-effectiveness, other nanomedicine-based methods also show promise in improving delivery efficiency and patient outcomes. Safety concerns, including potential toxicity and immunogenicity issues, must be addressed, with the FDA providing guidance for the safe development of nanotechnology-based products. Future directions in nanomedicine will focus on creating next-generation nanocarriers with precise targeting, real-time monitoring, and stimuli-responsive features to optimize diabetes treatment outcomes and patient safety. This review delves into the current state of nanomedicine for insulin delivery, examining various types of nanocarriers and their mechanisms of action, and discussing the challenges and future directions in developing safe and effective nanomedicine-based therapies for diabetes management.
Collapse
Affiliation(s)
- Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Roberto Nilo
- Data Collection G-STeP Research Core Facility, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - Davide Nilo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Vincenzo Russo
- Department of Biology, College of Science and Technology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
- Division of Cardiology, Department of Medical Translational Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | | | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| |
Collapse
|
21
|
Zhao F, Wang J, Zhang Y, Hu J, Li C, Liu S, Li R, Du R. In vivo Fate of Targeted Drug Delivery Carriers. Int J Nanomedicine 2024; 19:6895-6929. [PMID: 39005963 PMCID: PMC11246094 DOI: 10.2147/ijn.s465959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
This review aimed to systematically investigate the intracellular and subcellular fate of various types of targeting carriers. Upon entering the body via intravenous injection or other routes, a targeting carrier that can deliver therapeutic agents initiates their journey. If administered intravenously, the carrier initially faces challenges presented by the blood circulation before reaching specific tissues and interacting with cells within the tissue. At the subcellular level, the car2rier undergoes processes, such as drug release, degradation, and metabolism, through specific pathways. While studies on the fate of 13 types of carriers have been relatively conclusive, these studies are incomplete and lack a comprehensive analysis. Furthermore, there are still carriers whose fate remains unclear, underscoring the need for continuous research. This study highlights the importance of comprehending the in vivo and intracellular fate of targeting carriers and provides valuable insights into the operational mechanisms of different carriers within the body. By doing so, researchers can effectively select appropriate carriers and enhance the successful clinical translation of new formulations.
Collapse
Affiliation(s)
- Fan Zhao
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Jitong Wang
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Yu Zhang
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Jinru Hu
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Chenyang Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Shuainan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Key Laboratory of Polymorphic Drugs of Beijing, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
- Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Ruixiang Li
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| | - Ruofei Du
- Engineering Research Center of Modern Preparation Technology of TCM, Ministry of Education, Shanghai, 201203, People’s Republic of China
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People’s Republic of China
| |
Collapse
|
22
|
Baghbanbashi M, Shiran HS, Kakkar A, Pazuki G, Ristroph K. Recent advances in drug delivery applications of aqueous two-phase systems. PNAS NEXUS 2024; 3:pgae255. [PMID: 39006476 PMCID: PMC11245733 DOI: 10.1093/pnasnexus/pgae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 07/16/2024]
Abstract
Aqueous two-phase systems (ATPSs) are liquid-liquid equilibria between two aqueous phases that usually contain over 70% water content each, which results in a nontoxic organic solvent-free environment for biological compounds and biomolecules. ATPSs have attracted significant interest in applications for formulating carriers (microparticles, nanoparticles, hydrogels, and polymersomes) which can be prepared using the spontaneous phase separation of ATPSs as a driving force, and loaded with a wide range of bioactive materials, including small molecule drugs, proteins, and cells, for delivery applications. This review provides a detailed analysis of various ATPSs, including strategies employed for particle formation, polymerization of droplets in ATPSs, phase-guided block copolymer assemblies, and stimulus-responsive carriers. Processes for loading various bioactive payloads are discussed, and applications of these systems for drug delivery are summarized and discussed.
Collapse
Affiliation(s)
- Mojhdeh Baghbanbashi
- Department of Agricultural and Biological Engineering, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907, USA
| | - Hadi Shaker Shiran
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran 1591634311, Iran
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke St West, Montreal, QC H3A 0B8, Canada
| | - Gholamreza Pazuki
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran 1591634311, Iran
| | - Kurt Ristroph
- Department of Agricultural and Biological Engineering, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907, USA
| |
Collapse
|
23
|
Hofmann D, Sychev D, Zagradska-Paromova Z, Bittrich E, Auernhammer GK, Gaitzsch J. Surface Topology of Redox- and Thermoresponsive Nanogel Droplets. Macromol Rapid Commun 2024; 45:e2400049. [PMID: 38685191 DOI: 10.1002/marc.202400049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/27/2024] [Indexed: 05/02/2024]
Abstract
Hydrogels are usually depicted as a homogenous polymer block with a distinct surface. While defects in the polymer structure are looked into frequently, structural irregularities on the hydrogel surface are often neglected. In this work, thin hydrogel layers of ≈100 nm thickness (nanogels) are synthesized and characterized for their structural irregularities, as they represent the surface of macrogels. The nanogels contain a main-chain responsiveness (thermo responsive) and a responsiveness in the cross-linking points (redox responsive). By combining data from ellipsometry using box-model and two-segment-model analysis, as well as atomic force microscopy, a more defined model of the nanogel surface can be developed. Starting with a more densely cross-linked network at the silica wafer surface, the density of cross-linking gradually decreases toward the hydrogel-solvent interface. Thermo-responsive behavior of the main chain affects the entire network equally as all chain segments change solubility. Cross-linker-based redox-responsiveness, on the other hand, is only governed by the inner, more cross-linked layers of the network. Such dual responsive nanogels hence allow for developing a more detailed model of a hydrogel surface from free radical polymerization. It provides a better understanding of structural defects in hydrogels and how they are affected by responsive functionalities.
Collapse
Affiliation(s)
- Doreen Hofmann
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069, Dresden, Germany
| | - Dmitrii Sychev
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
- Chair of Physical Chemistry of Polymeric Materials, Technische Universität Dresden, 01069, Dresden, Germany
| | - Zlata Zagradska-Paromova
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01069, Dresden, Germany
| | - Eva Bittrich
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
| | - Günter K Auernhammer
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
| | - Jens Gaitzsch
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069, Dresden, Germany
| |
Collapse
|
24
|
Qin H, Teng Y, Dai R, Wang A, Liu J. Glycan-based scaffolds and nanoparticles as drug delivery system in cancer therapy. Front Immunol 2024; 15:1395187. [PMID: 38799466 PMCID: PMC11116596 DOI: 10.3389/fimmu.2024.1395187] [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: 03/03/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Glycan-based scaffolds are unique in their high specificity, versatility, low immunogenicity, and ability to mimic natural carbohydrates, making them attractive candidates for use in cancer treatment. These scaffolds are made up of glycans, which are biopolymers with well biocompatibility in the human body that can be used for drug delivery. The versatility of glycan-based scaffolds allows for the modulation of drug activity and targeted delivery to specific cells or tissues, which increases the potency of drugs and reduces side effects. Despite their promise, there are still technical challenges in the design and production of glycan-based scaffolds, as well as limitations in their therapeutic efficacy and specificity.
Collapse
Affiliation(s)
- Henan Qin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yibin Teng
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Rui Dai
- Department of Pharmacy, Peking Union Medical University Hospital, Beijing, China
| | - Aman Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
25
|
Neamtu I, Ghilan A, Rusu AG, Nita LE, Chiriac VM, Chiriac AP. Design and applications of polymer-like peptides in biomedical nanogels. Expert Opin Drug Deliv 2024; 21:713-734. [PMID: 38916156 DOI: 10.1080/17425247.2024.2364651] [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: 01/31/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION Polymer nanogels are among the most promising nanoplatforms for use in biomedical applications. The substantial interest for these drug carriers is to enhance the transportation of bioactive substances, reduce the side effects, and achieve optimal action on the curative sites by targeting delivery and triggering the release of the drugs in a controlled and continuous mode. AREA COVERED The review discusses the opportunities, applications, and challenges of synthetic polypeptide nanogels in biomedicine, with an emphasis on the recent progress in cancer therapy. It is evidenced by the development of polypeptide nanogels for better controlled drug delivery and release, in complex in vivo microenvironments in biomedical applications. EXPERT OPINION Polypeptide nanogels can be developed by choosing the amino acids from the peptide structure that are suitable for the type of application. Using a stimulus - sensitive peptide nanogel, it is possible to obtain the appropriate transport and release of the drug, as well as to achieve desirable therapeutic effects, including safety, specificity, and efficiency. The final system represents an innovative way for local and sustained drug delivery at a specific site of the body.
Collapse
Affiliation(s)
- Iordana Neamtu
- Natural Polymers, Bioactive and Biocompatible Materials Laboratory, Petru Poni Institute of Macromolecular Chemistry, Iasi, Romania
| | - Alina Ghilan
- Natural Polymers, Bioactive and Biocompatible Materials Laboratory, Petru Poni Institute of Macromolecular Chemistry, Iasi, Romania
| | - Alina Gabriela Rusu
- Natural Polymers, Bioactive and Biocompatible Materials Laboratory, Petru Poni Institute of Macromolecular Chemistry, Iasi, Romania
| | - Loredana Elena Nita
- Natural Polymers, Bioactive and Biocompatible Materials Laboratory, Petru Poni Institute of Macromolecular Chemistry, Iasi, Romania
| | - Vlad Mihai Chiriac
- Faculty of Electronics Telecommunications and Information Technology, Gh. Asachi Technical University, Iaşi, Romania
| | - Aurica P Chiriac
- Natural Polymers, Bioactive and Biocompatible Materials Laboratory, Petru Poni Institute of Macromolecular Chemistry, Iasi, Romania
| |
Collapse
|
26
|
Hashim PK, Abdrabou SSMA. Sub-100 nm carriers by template polymerization for drug delivery applications. NANOSCALE HORIZONS 2024; 9:693-707. [PMID: 38497369 DOI: 10.1039/d3nh00491k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Size-controlled drug delivery systems (DDSs) have gained significant attention in the field of pharmaceutical sciences due to their potential to enhance drug efficacy, minimize side effects, and improve patient compliance. This review provides a concise overview of the preparation method, advancements, and applications of size-controlled drug delivery systems focusing on the sub-100 nm size DDSs. The importance of tailoring the size for achieving therapeutic goals is briefly mentioned. We highlight the concept of "template polymerization", a well-established method in covalent polymerization that offers precise control over molecular weight. We demonstrate the utility of this approach in crafting a monolayer of a polymer around biomolecule templates such as DNA, RNA, and protein, achieving the generation of DDSs with sizes ranging from several tens of nanometers. A few representative examples of small-size DDSs that share a conceptual similarity to "template polymerization" are also discussed. This review concludes by briefly discussing the drug release behaviors and the future prospects of "template polymerization" for the development of innovative size-controlled drug delivery systems, which promise to optimize drug delivery precision, efficacy, and safety.
Collapse
Affiliation(s)
- P K Hashim
- Research Institute for Electronic Science, Hokkaido University, Kita 20, Nishi 10, Kita-ku, Sapporo, Hokkaido 001-0020, Japan.
- Graduate School of Life Science, Hokkaido University, Kita 10, Nishi 8, Kita-ku, Sapporo, Hokkaido 060-0810, Japan
| | | |
Collapse
|
27
|
Yugatama A, Huang YL, Hsu MJ, Lin JP, Chao FC, Lam JKW, Hsieh CM. Oral Delivery of Photopolymerizable Nanogels Loaded with Gemcitabine for Pancreatic Cancer Therapy: Formulation Design, and in vitro and in vivo Evaluations. Int J Nanomedicine 2024; 19:3753-3772. [PMID: 38686338 PMCID: PMC11057685 DOI: 10.2147/ijn.s443610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/19/2024] [Indexed: 05/02/2024] Open
Abstract
Background Gemcitabine (GEM) faces challenges of poor oral bioavailability and extensive first-pass metabolism. Currently, only injectable formulations are available for clinical use. Hence, there is an urgent demand for the development of advanced, efficacious, and user-friendly dosage forms to maintain its status as the primary treatment for pancreatic ductal adenocarcinoma (PDAC). Nanogels (NGs) offer a novel oral drug delivery system, ideal for hydrophilic compounds like GEM. This study aims to develop NGs tailored for GEM delivery, with the goal of enhancing cellular uptake and gastrointestinal permeability for improved administration in PDAC patients. Methods We developed cross-linked NGs via photopolymerization of methacryloyl for drug delivery of GEM. We reveal characterization, cytotoxicity, and cellular uptake studies in Caco-2 and MIA PaCa-2 cells. In addition, studies of in vitro permeability and pharmacokinetics were carried out to evaluate the bioavailability of the drug. Results Our results show NGs, formed via photopolymerization of methacryloyl, had a spherical shape with a size of 233.91±7.75 nm. Gemcitabine-loaded NGs (NGs-GEM) with 5% GelMA exhibited efficient drug loading (particle size: 244.07±19.52 nm). In vitro drug release from NGs-GEM was slower at pH 1.2 than pH 6.8. Cellular uptake studies indicated significantly enhanced uptake in both MIA PaCa-2 and Caco-2 cells. While there was no significant difference in GEM's AUC and Cmax between NGs-GEM and free-GEM groups, NGs-GEM showed markedly lower dFdU content (10.07 hr∙μg/mL) compared to oral free-GEM (19.04 hr∙μg/mL) after oral administration (p<0.01), highlighting NGs' efficacy in impeding rapid drug metabolism and enhancing retention. Conclusion In summary, NGs enhance cellular uptake, inhibit rapid metabolic degradation of GEM, and prolong retention after oral administration. These findings suggest NGs-GEM as a promising candidate for clinical use in oral pancreatic cancer therapy.
Collapse
Affiliation(s)
- Adi Yugatama
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Pharmacy, Sebelas Maret University, Surakarta, 57126, Indonesia
| | - Ya-Lin Huang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ming-Jen Hsu
- Department of Pharmacology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Jia-Pei Lin
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Fang-Ching Chao
- CNRS UMR 8612, Institut Galien Paris-Saclay, Université Paris-Saclay, Orsay, 91400, France
| | - Jenny K W Lam
- Department of Pharmaceutics, School of Pharmacy, University College, London, WC1N 1AX, UK
| | - Chien-Ming Hsieh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Pharmaceutics, School of Pharmacy, University College, London, WC1N 1AX, UK
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| |
Collapse
|
28
|
Du R, Fielding LA. pH-Responsive Nanogels Generated by Polymerization-Induced Self-Assembly of a Succinate-Functional Monomer. Macromolecules 2024; 57:3496-3501. [PMID: 38681060 PMCID: PMC11044572 DOI: 10.1021/acs.macromol.4c00427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 05/01/2024]
Abstract
Colloidal nanogels formed from a pH-responsive poly(succinate)-functional core and a poly(sulfonate)-functional corona were prepared via a previously unreported reversible addition-fragmentation chain-transfer (RAFT)-mediated aqueous emulsion polymerization-induced self-assembly (PISA) route. Specifically, a poly(potassium 3-sulfopropyl methacrylate) (PKSPMA50) macromolecular chain-transfer agent (macro-CTA) was synthesized via RAFT solution polymerization followed by chain-extension with a hydrophobic, carboxylic acid-functional, 2-(methacryloyloxy) ethyl succinate (MES) monomer at pH 2. Colloidal nanoparticles with tunable diameters between 66 to 150 nm, depending on the core composition, and narrow particle size distributions were obtained at 20% w/w solids. Well-defined pH-responsive nanogels that swell on increasing the pH could be prepared even without the addition of a cross-linking comonomer, and introducing an additional cross-linker to the core led to smaller nanogels with lower swelling ratios. These nanogels could reversibly change in size on cycling the pH between acidic and basic conditions and remain colloidally stable over a wide pH range and at 70 °C.
Collapse
Affiliation(s)
- Ruiling Du
- Department
of Materials, School of Natural Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
- Henry
Royce Institute, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| | - Lee A. Fielding
- Department
of Materials, School of Natural Sciences, University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
- Henry
Royce Institute, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| |
Collapse
|
29
|
Mali KK, Gavhane YN, Chakole RD. Natural Polymer-Based Nanogel for pH-Responsive Delivery of Sorafenib Tosylate in Hemangiosarcoma. AAPS PharmSciTech 2024; 25:83. [PMID: 38605211 DOI: 10.1208/s12249-024-02797-8] [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: 12/03/2023] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Smart nanomedicinal treatment for cancer manifests a solubility challenge with inherent nanoscale size and nonspecific release with stimuli-responsive potential. This is the limelight in novel chemotherapy to pursue physiochemical differences between the tumor microenvironment (TME) and normal cells, which introduces active groups of nanocarriers responding to various stimuli, endowing them with concise responses to various tumor-related signals. The nanogels were successfully prepared by a modified solvent evaporation technique. Nine batches were formulated by changing the chitosan concentration (12, 14, 16 mg/ml) and sonication time (5, 10, 15 min). The formulations were optimized for particle size and zeta potential with high percent entrapment efficiency (%EE) through Central Composite Design software. The optimized batch F7 had a 182-nm size and high zeta potential (64.5 mV) with 98% EE. The drug release of F7 was higher at pH 6 (97.556%) than at pH 7.4 (45.113%). The pharmacokinetic study shows that the release follows the Hixon plot model (R2 = 0.9334) that shifts to zero order (R2 = 0.9149). The nanogel F7 was observed for stability and showed an absence of color change, phase separation, and opacity for 6 months. In the present study, the pH difference between cancer cells and normal cells is the key point of the smart nanogel. This study is promising but challenging depending on the in vivo study. The nanogel was successfully prepared and evaluated for pH-responsive release. As hemangiosarcoma commonly occurs in dogs, this formulation helps to limit the difficulties with administration.
Collapse
Affiliation(s)
- Kiran K Mali
- Department of Pharmaceutics, Government College of Pharmacy, Karad, Shivaji University, Kolhapur, Maharashtra, 415124, India.
- Department of Pharmaceutics, Krishna Foundation's, Jaywant Institute of Pharmacy, Wathar, DBATU, Lonere, Karad, Maharashtra, 415139, India.
| | - Yogeshkumar N Gavhane
- Department of Pharmaceutics, Government College of Pharmacy, Karad, Shivaji University, Kolhapur, Maharashtra, 415124, India
| | - Rita D Chakole
- Department of Pharmacy, Government College of Pharmacy, Karad, Shivaji University, Kolhapur, Maharashtra, 415124, India
| |
Collapse
|
30
|
Yuniarsih N, Chaerunisaa AY, Elamin KM, Wathoni N. Polymeric Nanohydrogel in Topical Drug Delivery System. Int J Nanomedicine 2024; 19:2733-2754. [PMID: 38505165 PMCID: PMC10950079 DOI: 10.2147/ijn.s442123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 02/15/2024] [Indexed: 03/21/2024] Open
Abstract
Nanohydrogels (NH) are biodegradable polymers that have been extensively studied and utilized for various biomedical applications. Drugs in a topical medication are absorbed via the skin and carried to the intended location, where they are metabolized and eliminated from the body. With a focus on their pertinent contemporary treatments, this review aims to give a complete overview of recent advances in the creation and application of polymer NH in biomedicine. We will explore the key features that have driven advances in nanotechnology and discuss the significance of nanohydrogel-based formulations as vehicles for delivering therapeutic agents topically. The review will also cover the latest findings and references from the literature to support the advancements in nanotechnological technology related to the preparation and application of NH. In addition, we will also discuss the unique properties and potential applications of NH as drug delivery systems (DDS) for skin applications, underscoring their potential for effective topical therapeutic delivery. The challenge lies in efficiently delivering drugs through the skin's barrier to specific areas with high control. Environmentally sensitive systems, like polymer-based NH, show promise in treating dermatological conditions. Polymers are pivotal in developing these drug delivery systems, with NH offering advantages such as versatile drug loading, controlled release, and enhanced skin penetration.
Collapse
Affiliation(s)
- Nia Yuniarsih
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universitas Buana Perjuangan Karawang, Karawang, 41361, Indonesia
| | - Anis Yohana Chaerunisaa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Khaled M Elamin
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| |
Collapse
|
31
|
Huang R, Hirschbiegel CM, Lehot V, Liu L, Cicek YA, Rotello VM. Modular Fabrication of Bioorthogonal Nanozymes for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2300943. [PMID: 37042795 PMCID: PMC11234510 DOI: 10.1002/adma.202300943] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/21/2023] [Indexed: 06/19/2023]
Abstract
The incorporation of transition metal catalysts (TMCs) into nanoscaffolds generates nanocatalysts that replicate key aspects of enzymatic behavior. The TMCs can access bioorthogonal chemistry unavailable to living systems. These bioorthogonal nanozymes can be employed as in situ "factories" for generating bioactive molecules where needed. The generation of effective bioorthogonal nanozymes requires co-engineering of the TMC and the nanometric scaffold. This review presents an overview of recent advances in the field of bioorthogonal nanozymes, focusing on modular design aspects of both nanomaterial and catalyst and how they synergistically work together for in situ uncaging of imaging and therapeutic agents.
Collapse
Affiliation(s)
- Rui Huang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Cristina-Maria Hirschbiegel
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Victor Lehot
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Liang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Yagiz Anil Cicek
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA, 01003, USA
| |
Collapse
|
32
|
Dumitru MV, Neagu AL, Miron A, Roque MI, Durães L, Gavrilă AM, Sarbu A, Iovu H, Chiriac AL, Iordache TV. Retention of Ciprofloxacin and Carbamazepine from Aqueous Solutions Using Chitosan-Based Cryostructured Composites. Polymers (Basel) 2024; 16:639. [PMID: 38475322 DOI: 10.3390/polym16050639] [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: 01/16/2024] [Revised: 02/07/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Water pollution is becoming a great concern at the global level due to highly polluted effluents, which are charged year by year with increasing amounts of organic residues, dyes, pharmaceuticals and heavy metals. For some of these pollutants, the industrial treatment of wastewater is still relevant. Yet, in some cases, such as pharmaceuticals, specific treatment schemes are urgently required. Therefore, the present study describes the synthesis and evaluation of promising cryostructured composite adsorbents based on chitosan containing native minerals and two types of reinforcement materials (functionalized kaolin and synthetic silicate microparticles). The targeted pharmaceuticals refer to the ciprofloxacin (CIP) antibiotic and the carbamazepine (CBZ) drug, for which the current water treatment process seem to be less efficient, making them appear in exceedingly high concentrations, even in tap water. The study reveals first the progress made for improving the mechanical stability and resilience to water disintegration, as a function of pH, of chitosan-based cryostructures. Further on, a retention study shows that both pharmaceuticals are retained with high efficiency (up to 85.94% CIP and 86.38% CBZ) from diluted aqueous solutions.
Collapse
Affiliation(s)
- Marinela-Victoria Dumitru
- National Institute for Research & Development in Chemistry and Petrochemistry-ICECHIM, 202 Spl. Independenței, 060021 Bucharest, Romania
- Faculty of Chemical Engineering and Biotechnology, University POLITEHNICA of Bucharest, 1-7 Ghe. Polizu Street, 011061 Bucharest, Romania
| | - Ana-Lorena Neagu
- National Institute for Research & Development in Chemistry and Petrochemistry-ICECHIM, 202 Spl. Independenței, 060021 Bucharest, Romania
| | - Andreea Miron
- National Institute for Research & Development in Chemistry and Petrochemistry-ICECHIM, 202 Spl. Independenței, 060021 Bucharest, Romania
| | - Maria Inês Roque
- University of Coimbra, CERES-Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, Rua Silvio Lima, 3030-790 Coimbra, Portugal
| | - Luisa Durães
- University of Coimbra, CERES-Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, Rua Silvio Lima, 3030-790 Coimbra, Portugal
| | - Ana-Mihaela Gavrilă
- National Institute for Research & Development in Chemistry and Petrochemistry-ICECHIM, 202 Spl. Independenței, 060021 Bucharest, Romania
| | - Andrei Sarbu
- National Institute for Research & Development in Chemistry and Petrochemistry-ICECHIM, 202 Spl. Independenței, 060021 Bucharest, Romania
| | - Horia Iovu
- Faculty of Chemical Engineering and Biotechnology, University POLITEHNICA of Bucharest, 1-7 Ghe. Polizu Street, 011061 Bucharest, Romania
| | - Anita-Laura Chiriac
- National Institute for Research & Development in Chemistry and Petrochemistry-ICECHIM, 202 Spl. Independenței, 060021 Bucharest, Romania
| | - Tanța Verona Iordache
- National Institute for Research & Development in Chemistry and Petrochemistry-ICECHIM, 202 Spl. Independenței, 060021 Bucharest, Romania
| |
Collapse
|
33
|
Feng Q, Huo C, Wang M, Huang H, Zheng X, Xie M. Research progress on cuproptosis in cancer. Front Pharmacol 2024; 15:1290592. [PMID: 38357312 PMCID: PMC10864558 DOI: 10.3389/fphar.2024.1290592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Cuproptosis is a recently discovered form of cell death that is mediated by copper (Cu) and is a non-apoptotic form of cell death related to oligomerization of lipoylated proteins and loss of Fe-S protein clusters. Since its discovery, cuproptosis has been extensively studied by researchers for its mechanism and potential applications in the treatment of cancer. Therefore, this article reviews the specific mechanism of cuproptosis currently studied, as well as its principles and strategies for use in anti-cancer treatment, with the aim of providing a reference for cuproptosis-based cancer therapy.
Collapse
Affiliation(s)
- Qingbo Feng
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Chenyu Huo
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Maijian Wang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Handong Huang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xingbin Zheng
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ming Xie
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
34
|
Rana P, Singh C, Kaushik A, Saleem S, Kumar A. Recent advances in stimuli-responsive tailored nanogels for cancer therapy; from bench to personalized treatment. J Mater Chem B 2024; 12:382-412. [PMID: 38095136 DOI: 10.1039/d3tb02650g] [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: 12/21/2023]
Abstract
To improve the quality of health in a personalized manner, better control over pharmacologically relevant cargo formulation, organ-specific targeted delivery, and on-demand release of therapeutic agents is crucial. Significant work has been put into designing and developing revolutionary nanotherapeutics approaches for the effective monitoring and personalized treatment of disease. Nanogel (NG) has attracted significant interest because of its tremendous potential in cancer therapy and its environmental stimuli responsiveness. NG is considered a next-generation delivery technology due to its benefits like as size tunability, high loading, stimuli responsiveness, prolonged drug release via in situ gelling mechanisms, stability, and its potential to provide personalized therapy from the investigation of human genes and the genes in various types of cancers and its association with a selective anticancer drug. Stimuli-responsive NGs can be used as smart nanomedicines to detect and treat cancer and can be tuned as personalized medicine as well. This comprehensive review article's major objectives include the challenges of NGs' clinical translation for cancer treatment as well as its early preclinical successes and prospects.
Collapse
Affiliation(s)
- Prinsy Rana
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala-133207, Haryana, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Srinagar, Uttarakhand-246174, India
| | - Ajeet Kaushik
- NanoBiotech Lab, Department of Environmental Engineering, Florida Polytechnic University (FPU), Lakeland, FL, 33805-8531, USA
- School of Engineering, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Shakir Saleem
- Department of Public Health, College of Health Sciences, Saudi Electronic University, P. O. Box 93499, Riyadh 11673, Saudi Arabia
| | - Arun Kumar
- Department of Pharmacy, School of Health Sciences, Central University of South Bihar, Gaya-824209, India.
| |
Collapse
|
35
|
Yadav D, Sharma PK, Malviya R, Mishra PS, Surendra AV, Rao GSNK, Rani BR. Stimuli-responsive Biomaterials for Tissue Engineering Applications. Curr Pharm Biotechnol 2024; 25:981-999. [PMID: 37594093 DOI: 10.2174/1389201024666230818121821] [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: 01/24/2023] [Revised: 06/14/2023] [Accepted: 07/12/2023] [Indexed: 08/19/2023]
Abstract
The use of ''smart materials,'' or ''stimulus responsive'' materials, has proven useful in a variety of fields, including tissue engineering and medication delivery. Many factors, including temperature, pH, redox state, light, and magnetic fields, are being studied for their potential to affect a material's properties, interactions, structure, and/or dimensions. New tissue engineering and drug delivery methods are made possible by the ability of living systems to respond to both external stimuli and their own internal signals) for example, materials composed of stimuliresponsive polymers that self assemble or undergo phase transitions or morphology transformation. The researcher examines the potential of smart materials as controlled drug release vehicles in tissue engineering, aiming to enable the localized regeneration of injured tissue by delivering precisely dosed drugs at precisely timed intervals.
Collapse
Affiliation(s)
- Deepika Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Pramod Kumar Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Prem Shankar Mishra
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | | | - G S N Koteswara Rao
- Shobhaben Pratapbhai Patel School of Pharmacy, NMIMS Deemed University, Mumbai, India
| | - Budha Roja Rani
- Institute of Pharmaceutical Technology, Sri Padmavathi Mahila Visvavidyalayam, Tirupati, A.P., India
| |
Collapse
|
36
|
Mandal S, Vishvakarma P, Bhumika K. Developments in Emerging Topical Drug Delivery Systems for Ocular Disorders. Curr Drug Res Rev 2024; 16:251-267. [PMID: 38158868 DOI: 10.2174/0125899775266634231213044704] [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: 07/12/2023] [Revised: 10/23/2023] [Accepted: 11/10/2023] [Indexed: 01/03/2024]
Abstract
According to the current information, using nano gels in the eyes have therapeutic benefits. Industry growth in the pharmaceutical and healthcare sectors has been filled by nanotechnology. Traditional ocular preparations have a short retention duration and restricted drug bioavailability because of the eye's architectural and physiological barriers, a big issue for physicians, patients, and chemists. In contrast, nano gels can encapsulate drugs within threedimensional cross-linked polymeric networks. Because of their distinctive structural designs and preparation methods, they can deliver loaded medications in a controlled and sustained manner, enhancing patient compliance and therapeutic efficacy. Due to their excellent drugloading capacity and biocompatibility, nano-gels outperform other nano-carriers. This study focuses on using nano gels to treat eye diseases and provides a brief overview of their creation and response to stimuli. Our understanding of topical drug administration will be advanced using nano gel developments to treat common ocular diseases such as glaucoma, cataracts, dry eye syndrome, bacterial keratitis, and linked medication-loaded contact lenses and natural active ingredients.
Collapse
Affiliation(s)
- Suraj Mandal
- Department of Pharmacy, IIMT College of Medical Sciences, IIMT University, O-Pocket, Ganganagar, Meerut, 250001, U.P., India
| | - Prabhakar Vishvakarma
- Department of Pharmacy, IIMT College of Medical Sciences, IIMT University, O-Pocket, Ganganagar, Meerut, 250001, U.P., India
| | - Km Bhumika
- Department of Pharmacy, IIMT College of Medical Sciences, IIMT University, O-Pocket, Ganganagar, Meerut, 250001, U.P., India
| |
Collapse
|
37
|
Zhang L, Wang Z, Zhang R, Yang H, Wang WJ, Zhao Y, He W, Qiu Z, Wang D, Xiong Y, Zhao Z, Tang BZ. Multi-Stimuli-Responsive and Cell Membrane Camouflaged Aggregation-Induced Emission Nanogels for Precise Chemo-photothermal Synergistic Therapy of Tumors. ACS NANO 2023; 17:25205-25221. [PMID: 38091262 DOI: 10.1021/acsnano.3c08409] [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: 12/27/2023]
Abstract
Targeted and controllable drug release at lesion sites with the aid of visual navigation in real-time is of great significance for precise theranostics of cancers. Benefiting from the marvelous features (e.g., bright emission and phototheranostic effects in aggregates) of aggregation-induced emission (AIE) materials, constructing AIE-based multifunctional nanocarriers that act as all-arounders to integrate multimodalities for precise theranostics is highly desirable. Here, an intelligent nanoplatform (P-TN-Dox@CM) with homologous targeting, controllable drug release, and in vivo dual-modal imaging for precise chemo-photothermal synergistic therapy is proposed. AIE photothermic agent (TN) and anticancer drug (Dox) are encapsulated in thermo-/pH-responsive nanogels (PNA), and the tumor cell membranes are camouflaged onto the surface of nanogels. Active targeting can be realized through homologous effects derived from source tumor cell membranes, which advantageously elevates the specific drug delivery to tumor sites. After being engulfed into tumor cells, the nanogels exhibit a burst drug release at low pH. The near-infrared (NIR) photoinduced local hyperthermia can activate severe cytotoxicity and further accelerate drug release, thus generating enhanced synergistic chemo-photothermal therapy to thoroughly eradicate tumors. Moreover, P-TN-Dox@CM nanogels could achieve NIR-fluorescence/photothermal dual-modal imaging to monitor the dynamic distribution of therapeutics in real-time. This work highlights the great potential of smart P-TN-Dox@CM nanogels as a versatile nanoplatform to integrate multimodalities for precise chemo-photothermal synergistic therapy in combating cancers.
Collapse
Affiliation(s)
- Liping Zhang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, P. R. China
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P. R. China
| | - Zaiyu Wang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P. R. China
| | - Rongyuan Zhang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, P. R. China
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P. R. China
| | - Han Yang
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P. R. China
| | - Wen-Jin Wang
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P. R. China
| | - Yun Zhao
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P. R. China
| | - Wei He
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P. R. China
| | - Zijie Qiu
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P. R. China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Yu Xiong
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, P. R. China
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Zheng Zhao
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P. R. China
- HKUST-Shenzhen Research Institute, South Area Hi-Tech Park, Nanshan, Shenzhen, Guangdong 518057, P. R. China
| | - Ben Zhong Tang
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P. R. China
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong 999077, P. R. China
| |
Collapse
|
38
|
Jia X, Dou Z, Zhang Y, Li F, Xing B, Hu Z, Li X, Liu Z, Yang W, Liu Z. Smart Responsive and Controlled-Release Hydrogels for Chronic Wound Treatment. Pharmaceutics 2023; 15:2735. [PMID: 38140076 PMCID: PMC10747460 DOI: 10.3390/pharmaceutics15122735] [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: 10/24/2023] [Revised: 11/23/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Chronic wounds are a major health challenge that require new treatment strategies. Hydrogels are promising drug delivery systems for chronic wound healing because of their biocompatibility, hydration, and flexibility. However, conventional hydrogels cannot adapt to the dynamic and complex wound environment, which involves low pH, high levels of reactive oxygen species, and specific enzyme expression. Therefore, smart responsive hydrogels that can sense and respond to these stimuli are needed. Crucially, smart responsive hydrogels can modulate drug release and eliminate pathological factors by changing their properties or structures in response to internal or external stimuli, such as pH, enzymes, light, and electricity. These stimuli can also be used to trigger antibacterial responses, angiogenesis, and cell proliferation to enhance wound healing. In this review, we introduce the synthesis and principles of smart responsive hydrogels, describe their design and applications for chronic wound healing, and discuss their future development directions. We hope that this review will inspire the development of smart responsive hydrogels for chronic wound healing.
Collapse
Affiliation(s)
- Xintao Jia
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.J.); (Z.D.); (Y.Z.); (B.X.); (Z.H.); (X.L.); (Z.L.); (W.Y.)
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Zixuan Dou
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.J.); (Z.D.); (Y.Z.); (B.X.); (Z.H.); (X.L.); (Z.L.); (W.Y.)
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Ying Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.J.); (Z.D.); (Y.Z.); (B.X.); (Z.H.); (X.L.); (Z.L.); (W.Y.)
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Fanqin Li
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Bin Xing
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.J.); (Z.D.); (Y.Z.); (B.X.); (Z.H.); (X.L.); (Z.L.); (W.Y.)
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Zheming Hu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.J.); (Z.D.); (Y.Z.); (B.X.); (Z.H.); (X.L.); (Z.L.); (W.Y.)
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Xin Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.J.); (Z.D.); (Y.Z.); (B.X.); (Z.H.); (X.L.); (Z.L.); (W.Y.)
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Zhongyan Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.J.); (Z.D.); (Y.Z.); (B.X.); (Z.H.); (X.L.); (Z.L.); (W.Y.)
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Wenzhuo Yang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.J.); (Z.D.); (Y.Z.); (B.X.); (Z.H.); (X.L.); (Z.L.); (W.Y.)
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Zhidong Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (X.J.); (Z.D.); (Y.Z.); (B.X.); (Z.H.); (X.L.); (Z.L.); (W.Y.)
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
39
|
Zeng Y, Fan M, Zhou Q, Chen D, Jin T, Mu Z, Li L, Chen J, Qiu D, Zhang Y, Pan Y, Shen X, Cai X. Reactive Oxygen Species‐Activated CO Versatile Nanomedicine with Innate Gut Immune and Microbiome Remodeling Effects for Treating Inflammatory Bowel Disease. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202304381] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 01/06/2025]
Abstract
AbstractAbnormal activation of the gut mucosal immune system and a highly dysregulated gut microbiota play essential roles in the progression of inflammatory bowel disease (IBD). The clinical treatment of IBD remains highly challenging, with first‐line drugs showing limited efficacy and significant side effects. A reactive oxygen species (ROS)‐activated CO versatile nanomedicine (CMPs) capable of remodeling the gut immune‐microbiota microenvironment via potent anti‐oxidant, anti‐inflammatory, and antimicrobial effects is developed. CORM‐401‐loaded mannose‐modified peptide dendrimer nanogel: CMPs preferentially congregate on the surface of damaged colon mucosa after rectal administration and are subsequently internalized by activated immune cells. CORM‐401 can release numerous CO molecules in response to high ROS levels in cells and at the site of IBD, resulting in multiple therapeutic effects. In vitro and in vivo studies have demonstrated that CMPs scavenge ROS, suppress inflammatory responses, eliminate pathogens, and alleviate colitis in mouse models. RNA sequencing reveals that CMPs successfully remodel gut mucosal immune homeostasis by scavenging ROS, inhibiting NF‐κB/p38MAPK, activating PI3K‐Akt, and inhibiting HIF‐1‐induced glycolysis. 16S ribosomal RNA sequencing shows that CMPs can remodel the gut flora composition by restraining detrimental bacteria and augmenting beneficial bacteria. This study develops a promising and versatile nanomedicine for the management of IBD.
Collapse
Affiliation(s)
- Youyun Zeng
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Mengni Fan
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Qiang Zhou
- Ruian People's Hospital The Third Affiliated Hospital of Wenzhou Medical University Wenzhou 325016 China
| | - Dongfan Chen
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Ting Jin
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Zhixiang Mu
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Lin Li
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Jiale Chen
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Dongchao Qiu
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Yanmei Zhang
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Yihuai Pan
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| | - Xinkun Shen
- Ruian People's Hospital The Third Affiliated Hospital of Wenzhou Medical University Wenzhou 325016 China
| | - Xiaojun Cai
- School and Hospital of Stomatology Wenzhou Medical University Wenzhou 325027 China
| |
Collapse
|
40
|
Zhang R, Xu G, Su Y, Rao S. Potential Application of Ovalbumin Gel Nanoparticles Loaded with Carvacrol in the Preservation of Fresh Pork. Gels 2023; 9:941. [PMID: 38131927 PMCID: PMC10742687 DOI: 10.3390/gels9120941] [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: 11/06/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Plant essential oil has attracted much attention in delaying pork spoilage due to its safety, but its low antibacterial efficiency needs to be solved by encapsulation. Our previous research had fabricated a type of ovalbumin gel nanoparticles loaded with carvacrol (OCGn-2) using the gel-embedding method, which had a high encapsulation rate and antibacterial activity. The main purpose of this study was to further evaluate the stability and slow-release characteristics of OCGn-2 and potential quality effects of the nanoparticles on the preservation of fresh pork pieces during 4 °C storage. The particle test showed that the nanoparticles had better heat stability below 85 °C and salt stability below 90 mM. The in vitro release study indicated that the carvacrol in OCGn-2 followed a Fickian release mechanism. The pork preservation test suggested that the OCGn-2 coating treatments could remarkably restrict the quality decay of pork slices compared to free carvacrol or a physical mixture of ovalbumin and carvacrol treatment. Nano-encapsulation of ovalbumin is beneficial to the sustained release, enhanced oxidation resistance, and improved antibacterial activity of carvacrol. The study suggested that ovalbumin gel nanoparticles embedded with carvacrol could be applied as an efficient bacterial active packaging to extend the storage life of pork.
Collapse
Affiliation(s)
- Ruyi Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (R.Z.); (Y.S.)
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China;
| | - Guangwei Xu
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China;
| | - Yujie Su
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (R.Z.); (Y.S.)
| | - Shengqi Rao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (R.Z.); (Y.S.)
- College of Food Science and Engineering, Yangzhou University, Yangzhou 225127, China;
| |
Collapse
|
41
|
Jia J, Liu RK, Sun Q, Wang JX. Efficient Construction of pH-Stimuli-Responsive Colloidosomes with High Encapsulation Efficiency. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023. [PMID: 38015806 DOI: 10.1021/acs.langmuir.3c02415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Intelligent responsive colloidosomes have attracted increasing attention for their potential to enhance the efficacy and decrease the side effects of drugs in biomedical applications. However, a low encapsulation efficiency and complicated preparation method greatly limit their development. Herein, we report an efficient approach for the construction of pH-stimuli-responsive colloidosomes with high encapsulation efficiency by a high-gravity technology. The conditions under which latex particles with different methacrylic acid contents can successfully self-assemble into colloidosomes are explored. During the preparation process, emulsions emulsified for only 10 min at 2500 rpm in a unique high-gravity shearing surroundings are clarified owing to the greatly enhanced micromixing, while the emulsions emulsified for 30 min by a traditional high-speed shear machine at 4000 rpm are still yellow-white. More importantly, regular spherical colloidosomes encapsulating an anticancer drug doxorubicin not only achieve a small mean diameter of 2.82 μm but also realize a high encapsulation efficiency of 76.5%. The release performance of doxorubicin has an obvious pH-stimuli-responsive regularity and follows the first-order model of sustained release. The construction of intelligent responsive colloidosomes as drug carriers provides a route for controlled drug release and biomedical applications.
Collapse
Affiliation(s)
- Jia Jia
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, PR China
- Research Center of the Ministry of Education for High Gravity Engineering and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Rong-Kun Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, PR China
- Research Center of the Ministry of Education for High Gravity Engineering and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Qian Sun
- Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology and School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, PR China
| | - Jie-Xin Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, PR China
- Research Center of the Ministry of Education for High Gravity Engineering and Technology, Beijing University of Chemical Technology, Beijing 100029, PR China
| |
Collapse
|
42
|
Yang J, Shang J, Yang L, Wei D, Wang X, Deng Q, Zhong Z, Ye Y, Zhou M. Nanotechnology-Based Drug Delivery Systems for Honokiol: Enhancing Therapeutic Potential and Overcoming Limitations. Int J Nanomedicine 2023; 18:6639-6665. [PMID: 38026538 PMCID: PMC10656744 DOI: 10.2147/ijn.s431409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Honokiol (HNK) is a small-molecule polyphenol that has garnered considerable attention due to its diverse pharmacological properties, including antitumor, anti-inflammatory, anti-bacterial, and anti-obesity effects. However, its clinical application is restricted by challenges such as low solubility, poor bioavailability, and rapid metabolism. To overcome these limitations, researchers have developed a variety of nano-formulations for HNK delivery. These nano-formulations offer advantages such as enhanced solubility, improved bioavailability, extended circulation time, and targeted drug delivery. However, existing reviews of HNK primarily focus on its clinical and pharmacological features, leaving a gap in the comprehensive evaluation of HNK delivery systems based on nanotechnology. This paper aims to bridge this gap by comprehensively reviewing different types of nanomaterials used for HNK delivery over the past 15 years. These materials encompass vesicle delivery systems, nanoparticles, polymer micelles, nanogels, and various other nanocarriers. The paper details various HNK nano-delivery strategies and summarizes their latest applications, development prospects, and future challenges. To compile this review, we conducted an extensive search using keywords such as "honokiol", "nanotechnology", and "drug delivery system" on reputable databases, including PubMed, Scopus, and Web of Science, covering the period from 2008 to 2023. Through this search, we identified and selected approximately 90 articles that met our specific criteria.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Jinlu Shang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Liuxuan Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Daiqing Wei
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Xia Wang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Qinmin Deng
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Zhirong Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yun Ye
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
43
|
Marzini Irranca S, García Schejtman SD, Rosso AP, Coronado EA, Martinelli M. Hybrid nanogels by direct mixing of chitosan, tannic acid and magnetite nanoparticles: processes involved in their formation and potential catalytic properties. SOFT MATTER 2023; 19:8378-8385. [PMID: 37873678 DOI: 10.1039/d3sm00822c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Magnetite (Fe3O4) nanoparticles (MNPs) as nanocatalysts have drawn considerable attention because of their unique properties such as peroxidase-like activity. However, their biodistribution and availability for specific treatments still need to be improved. In this study, a simple and convenient strategy for the synthesis of hybrid nanogels (NGs) is described, which involves direct mixing of biomaterials such as chitosan (Ch) and tannic acid (TA), with the incorporation of MNPs, under oxidising conditions, using the inverse nanoemulsion method. The different processes involved in the formation of these hybrid nanosystems as well as their morphological and chemical structure are investigated using optical, spectroscopic, and electron microscopic techniques (DLS, UV-VIS, FT-IR, XPS, TEM, and SEM-EDS). It is demonstrated that ∼11 nm synthesized MNPs, post-functionalized with oxidised TA, act as covalent crosslinkers. As a proof of concept, the potential use of these materials in nanocatalytic medicine was evaluated using a colorimetric method based on the oxidation of 3,3',5,5'-tetramethylbenzidine (TMB) in hydrogen peroxide. The results show that these hybrid nanogels have the same peroxidase-like activity as bare MNPs, indicating that the organic nanostructure stabilises the inorganic nanoparticles without any significant change in the catalytic properties. Therefore, this kind of nanomaterial has promising potential for use in nanocatalytic medicine with improved biocompatibility and biodistribution.
Collapse
Affiliation(s)
- Santiago Marzini Irranca
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Orgánica, Materiales Poliméricos, Híbridos y Nanoarquitectónicos (LaMaP), Córdoba, Argentina.
- Instituto de Investigación y Desarrollo en Ingeniería de Procesos y Química Aplicada (IPQA), CONICET, Córdoba, Argentina.
| | - Sergio D García Schejtman
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Orgánica, Materiales Poliméricos, Híbridos y Nanoarquitectónicos (LaMaP), Córdoba, Argentina.
- Instituto de Investigación y Desarrollo en Ingeniería de Procesos y Química Aplicada (IPQA), CONICET, Córdoba, Argentina.
| | - Anabella P Rosso
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Orgánica, Materiales Poliméricos, Híbridos y Nanoarquitectónicos (LaMaP), Córdoba, Argentina.
- Instituto de Investigaciones en Físico-Química de Córdoba (INFIQC), CONICET, Córdoba, Argentina
| | - Eduardo A Coronado
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Fisicoquímica, Plasmónica Molecular, Bio(nanoplasmónica), Espectroscopías ultrasensibles, Córdoba, Argentina.
- Instituto de Investigaciones en Físico-Química de Córdoba (INFIQC), CONICET, Córdoba, Argentina
| | - Marisa Martinelli
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Orgánica, Materiales Poliméricos, Híbridos y Nanoarquitectónicos (LaMaP), Córdoba, Argentina.
- Instituto de Investigación y Desarrollo en Ingeniería de Procesos y Química Aplicada (IPQA), CONICET, Córdoba, Argentina.
| |
Collapse
|
44
|
Bakshi S, Pandey P, Mohammed Y, Wang J, Sailor MJ, Popat A, Parekh HS, Kumeria T. Porous silicon embedded in a thermoresponsive hydrogel for intranasal delivery of lipophilic drugs to treat rhinosinusitis. J Control Release 2023; 363:452-463. [PMID: 37769816 PMCID: PMC11484479 DOI: 10.1016/j.jconrel.2023.09.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/09/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Intranasal delivery is the most preferred route of drug administration for treatment of a range of nasal conditions including chronic rhinosinusitis (CRS), caused by an infection and inflammation of the nasal mucosa. However, localised delivery of lipophilic drugs for persistent nasal inflammation is a challenge especially with traditional topical nasal sprays. In this study, a composite thermoresponsive hydrogel is developed and tuned to obtain desired rheological and physiochemical properties suitable for intranasal administration of lipophilic drugs. The composite is comprised of drug-loaded porous silicon (pSi) particles embedded in a poloxamer 407 (P407) hydrogel matrix. Mometasone Furoate (MF), a lipophilic corticosteroid (log P of 4.11), is used as the drug, which is loaded onto pSi particles at a loading capacity of 28 wt%. The MF-loaded pSi particles (MF@pSi) are incorporated into the P407-based thermoresponsive hydrogel (HG) matrix to form the composite hydrogel (MF@pSi-HG) with a final drug content ranging between 0.1 wt% to 0.5 wt%. Rheomechanical studies indicate that the MF@pSi component exerts a minimal impact on gelation temperature or strength of the hydrogel host. The in-vitro release of the MF payload from MF@pSi-HG shows a pronounced increase in the amount of drug released over 8 h (4.5 to 21-fold) in comparison to controls consisting of pure MF incorporated in hydrogel (MF@HG), indicating an improvement in kinetic solubility of MF upon loading into pSi. Ex-vivo toxicity studies conducted on human nasal mucosal tissue show no adverse effect from exposure to either pure HG or the MF@pSi-HG formulation, even at the highest drug content of 0.5 wt%. Experiments on human nasal mucosal tissue show the MF@pSi-HG formulation deposits a quantity of MF into the tissues within 8 h that is >19 times greater than the MF@HG control (194 ± 7 μg of MF/g of tissue vs. <10 μg of MF/g of tissue, respectively).
Collapse
Affiliation(s)
- Shrishty Bakshi
- School of Pharmacy, The University of Queensland, Queensland 4102, Australia
| | - Preeti Pandey
- School of Pharmacy, The University of Queensland, Queensland 4102, Australia
| | - Yousuf Mohammed
- Therapeutics Research Group, Diamantina Institute, University of Queensland, Brisbane, Queensland 4102, Australia
| | - Joanna Wang
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA 94305, United States of America
| | - Michael J Sailor
- Department of Chemistry and Biochemistry, University of California-San Diego, La Jolla, CA 92093, United States of America
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Queensland 4102, Australia.
| | - Harendra S Parekh
- School of Pharmacy, The University of Queensland, Queensland 4102, Australia.
| | - Tushar Kumeria
- School of Pharmacy, The University of Queensland, Queensland 4102, Australia; School of Materials Science and Engineering, The University of New South Wales, New South Wales 2052, Australia; Australian Centre for Nanomedicine, The University of New South Wales, New South Wales 2052, Australia.
| |
Collapse
|
45
|
Zhao Q, Yue X, Miaomiao L, Yanming W, Wu G. Nano-injectable pH/NIR-responsive hydrogel for chemo-photothermal synergistic drug delivery. J Biomater Appl 2023; 38:614-628. [PMID: 37918422 DOI: 10.1177/08853282231209653] [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] [Indexed: 11/04/2023]
Abstract
Conventional cancer treatments are highly toxic and ineffective; therefore, it is essential to develop less toxic and minimally invasive treatment methods. A pH/Near Infra-red (NIR) dual-responsive, nano-injectable smart hydrogel was fabricated by incorporating CuS nanoparticles into the hydrogel networks formed by a random copolymer of N-isopropylacrylamide (NIPAM) and double-bond functionalized uracil. Microstructural characterizations of synthesized polymer and hydrogels were carried out using transmission electron microscope (TEM), scanning electron microscope (SEM), nuclear magnetic resonance (NMR) and fourier transform infrared spectroscopy (FT-IR). Multiple hydrogen bonding interactions between uracils function as physical cross-linking points to construct the network structure of the polymeric nanogel without the addition of additional cross-linking agents, ensuring the material's safety. The amino group on the structure of uracil gives the uracil-modified polymeric hydrogel excellent pH responsiveness. Notably, as a temperature-responsive material, poly (N-isopropylacrylamide) (PNIPAM) nanogel solution can achieve in situ gel formation (within 100 s at 37°C) above its lower critical solution temperature (LCST), granting injectability to polymeric solutions. Moreover, using a hierarchical construction strategy, the variable loading of DOX and CuS was achieved. First, a heterogeneous system was created by encapsulating doxorubicin (DOX) inside the nanogel via hydrophobic and π-π stacking interactions, followed by the introduction of CuS nanoparticles as photosensitizers outside of the nanogels. Due to the presence of CuS nanoparticles, the gel is able to convert NIR light into local heat to enhance the destruction of tumor cells while simultaneously achieving rapid in situ gel formation. The in situ-forming hydrogel showed promising tissue biocompatibility. The in vitro antitumor test demonstrated the capacity of the nanocomposite hydrogel for chemo-photothermal synergistic therapy. Therefore, this prepared platform has the potential to become a safe and effective, smart-responsive drug carrier for chemotherapy and PTT synergy, a minimally invasive material for tumor treatment.
Collapse
Affiliation(s)
- Qian Zhao
- Key Laboratory of Functional Polymer Materials of MOE, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, China
| | - Xu Yue
- Key Laboratory of Functional Polymer Materials of MOE, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, China
| | - Liu Miaomiao
- College of Pharmacy, Nankai University, Tianjin, China
| | - Wang Yanming
- College of Pharmacy, Nankai University, Tianjin, China
| | - Guolin Wu
- Key Laboratory of Functional Polymer Materials of MOE, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, China
| |
Collapse
|
46
|
Ashwani PV, Gopika G, Arun Krishna KV, Jose J, John F, George J. Stimuli-Responsive and Multifunctional Nanogels in Drug Delivery. Chem Biodivers 2023; 20:e202301009. [PMID: 37718283 DOI: 10.1002/cbdv.202301009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/19/2023]
Abstract
Nanogels represent promising drug delivery systems in the biomedical field, designed to overcome challenges associated with standard treatment approaches. Stimuli-responsive nanogels, often referred to as intelligent materials, have garnered significant attention for their potential to enhance control over properties such as drug release and targeting. Furthermore, researchers have recently explored the application of nanogels in diverse sectors beyond biomedicine including sensing materials, catalysts, or adsorbents for environmental applications. However, to fully harness their potential as practical delivery systems, further research is required to better understand their pharmacokinetic behaviour, interactions between nanogels and bio distributions, as well as toxicities. One promising future application of stimuli-responsive multifunctional nanogels is their use as delivery agents in cancer treatment, offering an alternative to overcome the challenges with conventional approaches. This review discusses various synthetic methods employed in developing nanogels as efficient carriers for drug delivery in cancer treatment. The investigations explore, the key aspects of nanogels, including their multifunctionality and stimuli-responsive properties, as well as associated toxicity concerns. The discussions presented herein aim to provide the readers a comprehensive understanding of the potential of nanogels as smart drug delivery systems in the context of cancer therapy.
Collapse
Affiliation(s)
- P V Ashwani
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - G Gopika
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - K V Arun Krishna
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Josena Jose
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Franklin John
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| | - Jinu George
- Bio-organic Laboratory, Department of Chemistry, Sacred Heart College, Kochi, 682013, India
| |
Collapse
|
47
|
Neumann-Tran TMP, López-Iglesias C, Navarro L, Quaas E, Achazi K, Biglione C, Klinger D. Poly( N-acryloylmorpholine) Nanogels as Promising Materials for Biomedical Applications: Low Protein Adhesion and High Colloidal Stability. ACS APPLIED POLYMER MATERIALS 2023; 5:7718-7732. [DOI: 10.1021/acsapm.3c00890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Thi Mai Phuong Neumann-Tran
- Institute of Pharmacy (Pharmaceutical Chemistry) Freie Universität Berlin, Königin-Luise -Str.2-4, Berlin 14195, Germany
| | - Clara López-Iglesias
- Institute of Pharmacy (Pharmaceutical Chemistry) Freie Universität Berlin, Königin-Luise -Str.2-4, Berlin 14195, Germany
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, I+D Farma Group (GI-1645), Faculty of Pharmacy, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Campus Vida s/n, 15782 Santiago de Compostela, Spain
| | - Lucila Navarro
- Institute of Pharmacy (Pharmaceutical Chemistry) Freie Universität Berlin, Königin-Luise -Str.2-4, Berlin 14195, Germany
| | - Elisa Quaas
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstrasse 23a, Berlin 14195, Germany
| | - Katharina Achazi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Altensteinstrasse 23a, Berlin 14195, Germany
| | - Catalina Biglione
- Institute of Pharmacy (Pharmaceutical Chemistry) Freie Universität Berlin, Königin-Luise -Str.2-4, Berlin 14195, Germany
- Advanced Porous Materials Unit (APMU), IMDEA Energy Institute, E-28935 Madrid, Spain
| | - Daniel Klinger
- Institute of Pharmacy (Pharmaceutical Chemistry) Freie Universität Berlin, Königin-Luise -Str.2-4, Berlin 14195, Germany
| |
Collapse
|
48
|
Duan QY, Zhu YX, Jia HR, Wang SH, Wu FG. Nanogels: Synthesis, properties, and recent biomedical applications. PROGRESS IN MATERIALS SCIENCE 2023; 139:101167. [DOI: 10.1016/j.pmatsci.2023.101167] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
49
|
WIBOWO AA, BÜTÜN V. pH-responsive intermediary layer cross-linked micelles from zwitterionic triblock copolymers and investigation of their drug-release behaviors. Turk J Chem 2023; 47:1103-1115. [PMID: 38173758 PMCID: PMC10760816 DOI: 10.55730/1300-0527.3597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 10/31/2023] [Accepted: 09/30/2023] [Indexed: 01/05/2024] Open
Abstract
ABC-type triblock copolymers, namely poly[(ethylene glycol)methyl ether]-block-poly(tert-butyl methacrylate)-block-poly[2-N-(diisopropylamino)ethyl methacrylate] (MPEG-b-PBuMA-b-PDPA), were first synthesized and then the middle blocks were successfully converted into poly(methacrylic acid) to obtain MPEG-b-PMAA-b-PDPA zwitterionic triblock copolymers. These block copolymers were soluble in water and formed micellar aggregates with complex cores via hydrogen bonding interactions between MPEG and PMAA blocks below pH 4.0. When the pH was between 5.0 and 7.0, due to charge compensation between partially protonated PDPA and partially ionized PMAA blocks, micelles with polyion complex cores were observed. If the solution pH was above 8.0, deprotonation of tertiary amine groups provided a hydrophobic character to the PDPA block, which resulted in the formation of PDPA-core micelles while MPEG/anionic PMAA hybrid blocks formed hydrated coronas. Intermediary layer cross-linked (ILCL) micelles from PDPA-core micelles were also prepared by cross-linking the inner PMAA shell. The hydrophobic drug dipyridamole (DIP) was used to investigate the release profile of ILCL micelles. DIP can be loaded to the PDPA cores of the micelles in basic aqueous media. An increase in the degree of cross-linking causes slower release for the model drug. It was concluded that the more complex matrix formation in the intermediary layer of the micelles via cross-linking retards the drug release from the core.
Collapse
Affiliation(s)
- Agung Ari WIBOWO
- Department of Polymer Science and Technology, Institute of Science, Eskişehir Osmangazi University, Meşelik Campus, Eskişehir,
Turkiye
| | - Vural BÜTÜN
- Department of Polymer Science and Technology, Institute of Science, Eskişehir Osmangazi University, Meşelik Campus, Eskişehir,
Turkiye
- Department of Chemistry, Faculty of Science, Eskişehir Osmangazi University, Eskişehir,
Turkiye
| |
Collapse
|
50
|
Xie G, Lin S, Wu F, Liu J. Nanomaterial-based ophthalmic drug delivery. Adv Drug Deliv Rev 2023; 200:115004. [PMID: 37433372 DOI: 10.1016/j.addr.2023.115004] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/13/2023]
Abstract
The low bioavailability and side effects of conventional drugs for eye disease necessitate the development of efficient drug delivery systems. Accompanying the developments of nanofabrication techniques, nanomaterials have been recognized as promising tools to overcome these challenges due to their flexible and programmable properties. Given the advances achieved in material science, a broad spectrum of functional nanomaterials capable of overcoming various ocular anterior and posterior segment barriers have been explored to satisfy the demands for ocular drug delivery. In this review, we first highlight the unique functions of nanomaterials suitable for carrying and transporting ocular drugs. Then, various functionalization strategies are emphasized to endow nanomaterials with superior performance in enhanced ophthalmic drug delivery. The rational design of several affecting factors is essential for ideal nanomaterial candidates and is depicted as well. Lastly, we introduce the current applications of nanomaterial-based delivery systems in the therapy of different ocular anterior and posterior segment diseases. The limitations of these delivery systems as well as potential solutions are also discussed. This work will inspire innovative design thinking for the development of nanotechnology-mediated strategies for advanced drug delivery and treatment toward ocular diseases.
Collapse
Affiliation(s)
- Guocheng Xie
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Feng Wu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|