1
|
Zhou Z, Han J, Lang P, Zhang M, Shu H, Zhang L, Huang S. ROS-responsive self-assembly nanoplatform overcomes hypoxia for enhanced photodynamic therapy. Biomater Sci 2024; 12:5105-5114. [PMID: 39221610 DOI: 10.1039/d4bm00712c] [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: 09/04/2024]
Abstract
Photodynamic therapy (PDT) has emerged as a promising treatment for malignant tumours in recent decades due to its impressive spatiotemporal selectivity, minimal invasiveness, and few adverse effects. Despite these advancements, there remain significant challenges in effectively delivering photosensitizers to tumours and overcoming tumour hypoxia to maximize the therapeutic benefits of PDT. Ongoing research efforts are focused on developing innovative strategies to overcome the above-mentioned challenges, such as nanoplatforms and combination therapy approaches. Hence, reactive oxygen species (ROS)-responsive polymeric micelles are promising candidates to enhance the distribution and retention of photosensitizers within tumours. Additionally, efforts to alleviate tumour hypoxia may further improve the anti-tumour effects of PDT. In this study, we designed ROS-responsive polymeric micelles (TC@PTP) co-loaded with a Tapp-COF, a porphyrin derivative, and capsaicin for PDT of melanoma. These ROS-responsive nanocarriers, constructed from thioketal (TK)-linked amphiphilic di-block copolymers (PEG5K-TK-PLGA5K), could accumulate in the tumor microenvironment and release drugs under the action of ROS. Capsaicin, acting as a biogenic respiratory inhibitor, suppressed mitochondrial respiration and the hypoxia-inducible factor 1 (HIF-1) signaling pathway, thereby increasing oxygen levels at the tumour site. These PDT-triggered ROS-responsive nanoparticles effectively alleviated the tumour hypoxic microenvironment and enhanced anti-tumour efficacy. With superior biocompatibility and tumour-targeting abilities, the platform holds great promise for advancing anti-tumour combination therapy.
Collapse
Affiliation(s)
- Zhaojie Zhou
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610000, China.
| | - Jiaxi Han
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610000, China.
| | - Puxin Lang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610000, China.
| | - Mengxing Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610000, China.
| | - Haozhou Shu
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610000, China.
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610000, China.
- Med-X Center for Materials, Sichuan University, Chengdu 610000, China
| | - Shiqi Huang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610000, China.
| |
Collapse
|
2
|
Mishra M, Verma R, Sharma A, Kumar K, Chawla R. Evaluation of Gemcitabine and Epigallocatechin-3-Gallate Loaded Solid Lipid Nanoparticles on Benzopyrene Induced Lung Cancer Model Via Intranasal Route: Improved Pharmacokinetics and Safety Profile. AAPS PharmSciTech 2024; 25:176. [PMID: 39085673 DOI: 10.1208/s12249-024-02892-w] [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: 05/08/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
The objective of this study was to create a new treatment for lung cancer using solid lipid nanoparticles (SLNs) loaded with gemcitabine (GEM) and epigallocatechin-3-gallate (EGCG) that can be administered through the nose. We analyzed the formulation for its effectiveness in terms of micromeritics, drug release, and anti-cancer activity in the benzopyrene-induced Swiss albino mice lung cancer model. We also assessed the pharmacokinetics, biodistribution, biocompatibility, and hemocompatibility of GEM-EGCG SLNs. The GEM-EGCG SLNs had an average particle size of 93.54 ± 11.02 nm, a polydispersity index of 0.146 ± 0.05, and a zeta potential of -34.7 ± 0.4 mV. The entrapment efficiency of GEM and EGCG was 93.39 ± 4.2% and 89.49 ± 5.1%, respectively, with a sustained release profile for both drugs. GEM-EGCG SLNs had better pharmacokinetics than other treatments, and a high drug targeting index value of 17.605 for GEM and 2.118 for EGCG, indicating their effectiveness in targeting the lungs. Blank SLNs showed no pathological lesions in the liver, kidney, and nasal region validating the safety of SLNs. GEM-EGCG SLNs also showed fewer pathological lesions than other treatments and a lower hemolysis rate of 1.62 ± 0.10%. These results suggest that GEM-EGCG SLNs could effectively treat lung cancer.
Collapse
Affiliation(s)
- Mohini Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Rinki Verma
- Department of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Aditya Sharma
- Sri Ganganagar Homoeopathic Medical College, Hospital & Research Center, Tantia University, Sri Ganganagar, Rajasthan, 335002, India
| | - Krishan Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Ruchi Chawla
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
3
|
Chen ZA, Wu CH, Wu SH, Huang CY, Mou CY, Wei KC, Yen Y, Chien IT, Runa S, Chen YP, Chen P. Receptor Ligand-Free Mesoporous Silica Nanoparticles: A Streamlined Strategy for Targeted Drug Delivery across the Blood-Brain Barrier. ACS NANO 2024; 18:12716-12736. [PMID: 38718220 PMCID: PMC11112986 DOI: 10.1021/acsnano.3c08993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/22/2024]
Abstract
Mesoporous silica nanoparticles (MSNs) represent a promising avenue for targeted brain tumor therapy. However, the blood-brain barrier (BBB) often presents a formidable obstacle to efficient drug delivery. This study introduces a ligand-free PEGylated MSN variant (RMSN25-PEG-TA) with a 25 nm size and a slight positive charge, which exhibits superior BBB penetration. Utilizing two-photon imaging, RMSN25-PEG-TA particles remained in circulation for over 24 h, indicating significant traversal beyond the cerebrovascular realm. Importantly, DOX@RMSN25-PEG-TA, our MSN loaded with doxorubicin (DOX), harnessed the enhanced permeability and retention (EPR) effect to achieve a 6-fold increase in brain accumulation compared to free DOX. In vivo evaluations confirmed the potent inhibition of orthotopic glioma growth by DOX@RMSN25-PEG-TA, extending survival rates in spontaneous brain tumor models by over 28% and offering an improved biosafety profile. Advanced LC-MS/MS investigations unveiled a distinctive protein corona surrounding RMSN25-PEG-TA, suggesting proteins such as apolipoprotein E and albumin could play pivotal roles in enabling its BBB penetration. Our results underscore the potential of ligand-free MSNs in treating brain tumors, which supports the development of future drug-nanoparticle design paradigms.
Collapse
Affiliation(s)
- Zih-An Chen
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Research
Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Cheng-Hsun Wu
- Nano
Targeting & Therapy Biopharma Inc., Taipei 10087, Taiwan
| | - Si-Han Wu
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International
Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chiung-Yin Huang
- Neuroscience
Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chung-Yuan Mou
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Nano
Targeting & Therapy Biopharma Inc., Taipei 10087, Taiwan
| | - Kuo-Chen Wei
- Neuroscience
Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department
of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- School
of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department
of Neurosurgery, New Taipei Municipal TuCheng
Hospital, New Taipei City 23652, Taiwan
| | - Yun Yen
- Center
for Cancer Translational Research, Tzu Chi
University, Hualien 970374, Taiwan
- Cancer
Center, Taipei Municipal WanFang Hospital, Taipei 116081, Taiwan
| | - I-Ting Chien
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Sabiha Runa
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- SRS Medical Communications,
LLC, Cleveland, Ohio 44124, United States
| | - Yi-Ping Chen
- Graduate
Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International
Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Peilin Chen
- Research
Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
4
|
Li X, Liu Y, Wu L, Zhao J. Molecular Nanoarchitectonics of Natural Photosensitizers and Their Derivatives Nanostructures for Improved Photodynamic Therapy. ChemMedChem 2024; 19:e202300599. [PMID: 38069595 DOI: 10.1002/cmdc.202300599] [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: 10/31/2023] [Revised: 12/01/2023] [Indexed: 01/25/2024]
Abstract
Natural photosensitizers (PSs) and their derivatives have drawn ever-increasing attention in photodynamic therapy (PDT) for their wild range of sources, desirable biocompatibility, and good photosensitivity. Nevertheless, many factors such as poor solubility, high body clearance rate, limited tumor targeting ability, and short excitation wavelengths severely hinder their applications in efficient PDT. In recent years, fabricating nanostructures by utilizing molecular assembly technique is proposed to solve these problems. This technique is easy to put into effect, and the assembled nanostructures could improve the physical properties of the PSs so as to meet the requirement of PDT. In this concept, we focus on the construction of natural PSs and their derivatives nanostructures through molecular assembly technique to enhance PDT efficacy (Figure 1). Furthermore, current challenges and future perspectives of natural PSs and their derivatives for efficient PDT are discussed.
Collapse
Affiliation(s)
- Xiaochen Li
- Shaanxi University of Chinese Medicine, 712046, Xianyang, China
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, 100029, Beijing, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Yilin Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS, Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Lili Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, 100029, Beijing, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Jie Zhao
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS, Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, China
| |
Collapse
|
5
|
Jin Z, Al Amili M, Guo S. Tumor Microenvironment-Responsive Drug Delivery Based on Polymeric Micelles for Precision Cancer Therapy: Strategies and Prospects. Biomedicines 2024; 12:417. [PMID: 38398021 PMCID: PMC10886702 DOI: 10.3390/biomedicines12020417] [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: 12/30/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
In clinical practice, drug therapy for cancer is still limited by its inefficiency and high toxicity. For precision therapy, various drug delivery systems, including polymeric micelles self-assembled from amphiphilic polymeric materials, have been developed to achieve tumor-targeting drug delivery. Considering the characteristics of the pathophysiological environment at the drug target site, the design, synthesis, or modification of environmentally responsive polymeric materials has become a crucial strategy for drug-targeted delivery. In comparison to the normal physiological environment, tumors possess a unique microenvironment, characterized by a low pH, high reactive oxygen species concentration, hypoxia, and distinct enzyme systems, providing various stimuli for the environmentally responsive design of polymeric micelles. Polymeric micelles with tumor microenvironment (TME)-responsive characteristics have shown significant improvement in precision therapy for cancer treatment. This review mainly outlines the most promising strategies available for exploiting the tumor microenvironment to construct internal stimulus-responsive drug delivery micelles that target tumors and achieve enhanced antitumor efficacy. In addition, the prospects of TME-responsive polymeric micelles for gene therapy and immunotherapy, the most popular current cancer treatments, are also discussed. TME-responsive drug delivery via polymeric micelles will be an efficient and robust approach for developing clinical cancer therapies in the future.
Collapse
Affiliation(s)
- Zhu Jin
- Correspondence: (Z.J.); (S.G.)
| | | | - Shengrong Guo
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China;
| |
Collapse
|
6
|
Zhang B, Liu H, Wang Y, Zhang Y, Cheng J. Application of singlet oxygen-activatable nanocarriers to boost X-ray-induced photodynamic therapy and cascaded ferroptosis for breast cancer treatment. J Mater Chem B 2023; 11:9685-9696. [PMID: 37789698 DOI: 10.1039/d3tb01887c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Ferroptosis has appealing antitumor potential that is mainly based on the accumulation of lipid peroxide to a lethal level. The cytotoxic singlet oxygen (1O2) generated from nanoscale X-ray-induced photodynamic therapy (X-PDT) may facilitate glutathione (GSH) depletion and further activate ferroptosis. To realize combined X-PDT and ferroptosis, a nanocarrier (D-NPVR) was engineered with a hyperbranched copolymer with 1O2-sensitive linkers, where both the photosensitizer (verteporfin) and ferroptosis inducer RAS-selective lethal small molecule 3 (RSL3) were encapsulated. Upon X-ray radiation, D-NPVR could produce a large amount of 1O2 for apoptosis. Subsequently, 1O2 triggered D-NP dissociation by cleavage of 1,2-bis(2-hydroxyethylthio)ethylene bonds to boost payload release and decrease levels of intracellular GSH via thiol oxidation. Liberated RSL3 is a covalent inhibitor for glutathione peroxide 4 (GPX4), which is responsible for detoxifying lipid peroxides to lipid alcohols with GSH assistance, and both 1O2-induced GSH depletion and GPX4 inactivation thereby produced ferroptotic cell death. Tumor growth inhibition in murine 4T1 tumor-bearing mice demonstrated that D-NPVR produced pronounced therapeutic efficiency where ferroptosis induction was supported by the GPX4 content and expression. This study highlights the contribution of 1O2-sensitive nanocarriers for promoting the potency of combined X-PDT and ferroptosis.
Collapse
Affiliation(s)
- Beibei Zhang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, P. R. China.
- Key Laboratory for Functional Magnetic resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou 450002, P. R. China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou 450002, P. R. China
| | - Hao Liu
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, P. R. China.
- Key Laboratory for Functional Magnetic resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou 450002, P. R. China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou 450002, P. R. China
| | - Yifei Wang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, P. R. China.
- Key Laboratory for Functional Magnetic resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou 450002, P. R. China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou 450002, P. R. China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, P. R. China.
- Key Laboratory for Functional Magnetic resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou 450002, P. R. China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou 450002, P. R. China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, P. R. China.
- Key Laboratory for Functional Magnetic resonance Imaging and Molecular Imaging of Henan Province, Zhengzhou 450002, P. R. China
- Engineering Research Center of Medical Imaging Intelligent Diagnosis and Treatment of Henan Province, Zhengzhou 450002, P. R. China
| |
Collapse
|
7
|
Aliabadi A, Hasannia M, Vakili-Azghandi M, Araste F, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. Synthesis approaches of amphiphilic copolymers for spherical micelle preparation: application in drug delivery. J Mater Chem B 2023; 11:9325-9368. [PMID: 37706425 DOI: 10.1039/d3tb01371e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
The formation of polymeric micelles in aqueous environments through the self-assembly of amphiphilic polymers can provide a versatile platform to increase the solubility and permeability of hydrophobic drugs and pave the way for their administration. In comparison to various self-assembly-based vehicles, polymeric micelles commonly have a smaller size, spherical morphology, and simpler scale up process. The use of polymer-based micelles for the encapsulation and carrying of therapeutics to the site of action triggered a line of research on the synthesis of various amphiphilic polymers in the past few decades. The extended knowledge on polymers includes biocompatible smart amphiphilic copolymers for the formation of micelles, therapeutics loading and response to external stimuli, micelles with a tunable drug release pattern, etc. Different strategies such as ring-opening polymerization, atom transfer radical polymerization, reversible addition-fragmentation chain-transfer, nitroxide mediated polymerization, and a combination of these methods were employed to synthesize copolymers with diverse compositions and topologies with the proficiency of self-assembly into well-defined micellar structures. The current review provides a summary of the important polymerization techniques and recent achievements in the field of drug delivery using micellar systems. This review proposes new visions for the design and synthesis of innovative potent amphiphilic polymers in order to benefit from their application in drug delivery fields.
Collapse
Affiliation(s)
- Ali Aliabadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Medicinal Chemistry Department, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maliheh Hasannia
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Masoume Vakili-Azghandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Fatemeh Araste
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Biotechnology Department, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Biotechnology Department, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Biotechnology Department, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Turhan Z, d’Arcy R, El Mohtadi F, Teixeira LI, Francini N, Geven M, Castagnola V, Alshamsan A, Benfenati F, Tirelli N. Dual Thermal- and Oxidation-Responsive Polymers Synthesized by a Sequential ROP-to-RAFT Procedure Inherently Temper Neuroinflammation. Biomacromolecules 2023; 24:4478-4493. [PMID: 36757736 PMCID: PMC10565819 DOI: 10.1021/acs.biomac.2c01365] [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: 11/16/2022] [Revised: 01/25/2023] [Indexed: 02/10/2023]
Abstract
This study is about multiple responsiveness in biomedical materials. This typically implies "orthogonality" (i.e., one response does not affect the other) or synergy (i.e., one increases efficacy or selectivity of the other), but an antagonist effect between responses may also occur. Here, we describe a family of very well-defined amphiphilic and micelle-forming block copolymers, which show both oxidative and temperature responses. They are produced via successive anionic ring-opening polymerization of episulfides and RAFT polymerization of dialkylacrylamides and differ only in the ratio between inert (N,N-dimethylacrylamide, DMA) and temperature-sensitive (N,N-diethylacrylamide, DEA) units. By scavenging Reactive Oxygen Species (ROS), these polymers are anti-inflammatory; through temperature responsiveness, they can macroscopically aggregate, which may allow them to form depots upon injection. The localization of the anti-inflammatory action is an example of synergy. An extensive evaluation of toxicity and anti-inflammatory effects on in vitro models, including BV2 microglia, C8D30 astrocytes and primary neurons, shows a link between capacity of aggregation and detrimental effects on viability which, albeit mild, can hinder the anti-inflammatory potential (antagonist action). Although limited in breadth (e.g., only in vitro models and only DEA as a temperature-responsive unit), this study suggests that single-responsive controls should be used to allow for a precise assessment of the (synergic or antagonist) potential of double-responsive systems.
Collapse
Affiliation(s)
- Zulfiye
Y. Turhan
- Laboratory
for Polymers and Biomaterials, Fondazione
Istituto Italiano di Tecnologia, 16163 Genova, Italy
- Division
of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United
Kingdom
| | - Richard d’Arcy
- Laboratory
for Polymers and Biomaterials, Fondazione
Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Farah El Mohtadi
- Division
of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United
Kingdom
| | - Lorena Infante Teixeira
- Laboratory
for Polymers and Biomaterials, Fondazione
Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Nora Francini
- Laboratory
for Polymers and Biomaterials, Fondazione
Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Mike Geven
- Laboratory
for Polymers and Biomaterials, Fondazione
Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Valentina Castagnola
- Center
for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132 Genova, Italy
- IRCCS
Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Aws Alshamsan
- Department
of Pharmaceutics, College of Pharmacy, King
Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
- Nanobiotechnology
Unit, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Fabio Benfenati
- Center
for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, 16132 Genova, Italy
- IRCCS
Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Nicola Tirelli
- Laboratory
for Polymers and Biomaterials, Fondazione
Istituto Italiano di Tecnologia, 16163 Genova, Italy
- Division
of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United
Kingdom
| |
Collapse
|
9
|
Chakraborty G, Meher M, Dash S, Rout RN, Pradhan S, Sahoo D. Strategies for Targeted Delivery via Structurally Variant Polymeric Nanocarriers. ChemistrySelect 2023; 8. [DOI: 10.1002/slct.202301626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/13/2023] [Indexed: 01/06/2025]
Abstract
AbstractThe last decade has seen a meteoric rise in studies investigating polymeric aggregates as nanocarriers. When it comes to morphology, size, functionality, and immunostability, polymeric nanocarriers (PNCs) are unparalleled. With characteristics such as large surface area to volume ratio, amphiphilic nano‐environment, non‐toxic components, chemically modifiable composition, external surface alteration potential, uniform particle size, and stimuli‐dependent self‐assembly, PNCs have emerged as strong candidates for therapeutic applications. The article reviews the latest research on different challenges and strategies for targeted drug delivery and shall serve as guide to the researchers in designing site‐specific nanocarriers for application in future. The review systematically discusses the fundamental structural variation of the nanocarriers with emphasis on the influence of chemical alterations and the resulting effects on functionality; addresses the difficulties encountered with modes of administration; target selectivity and stimulus response.
Collapse
Affiliation(s)
- Gulmi Chakraborty
- Department of Chemistry C.V. Raman Global University Odisha 752054 India
| | - Minakshi Meher
- Department of Chemistry C.V. Raman Global University Odisha 752054 India
| | - Sanjay Dash
- Department of Chemistry C.V. Raman Global University Odisha 752054 India
| | - Rudra Narayan Rout
- Department of Chemistry C.V. Raman Global University Odisha 752054 India
| | - Sibananda Pradhan
- Department of Chemistry C.V. Raman Global University Odisha 752054 India
| | - Dipanjali Sahoo
- Department of Chemistry C.V. Raman Global University Odisha 752054 India
| |
Collapse
|
10
|
Zhang Z, Feng J, Zhang T, Gao A, Sun C. Application of tumor pH/hypoxia-responsive nanoparticles for combined photodynamic therapy and hypoxia-activated chemotherapy. Front Bioeng Biotechnol 2023; 11:1197404. [PMID: 37362218 PMCID: PMC10289258 DOI: 10.3389/fbioe.2023.1197404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction: Cancer selectivity, including targeted internalization and accelerated drug release in tumor cells, remains a major challenge for designing novel stimuli-responsive nanocarriers to promote therapeutic efficacy. The hypoxic microenvironment created by photodynamic therapy (PDT) is believed to play a critical role in chemoresistance. Methods: We construct dual-responsive carriers (DANPCT) that encapsulate the photosensitizer chlorin e6 (Ce6) and hypoxia-activated prodrug tirapazamine (TPZ) to enable efficient PDT and PDT-boosted hypoxia-activated chemotherapy. Results and discussion: Due to TAT masking, DANPCT prolonged payload circulation in the bloodstream, and selective tumor cell uptake occurred via acidity-triggered TAT presentation. PDT was performed with a spatially controlled 660-nm laser to enable precise cell killing and exacerbate hypoxia. Hypoxia-responsive conversion of the hydrophobic NI moiety led to the disassembly of DANPCT, facilitating TPZ release. TPZ was reduced to cytotoxic radicals under hypoxic conditions, contributing to the chemotherapeutic cascade. This work offers a sophisticated strategy for programmed chemo-PDT.
Collapse
Affiliation(s)
- Zhang Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Jintang Feng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Tianzhu Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - An Gao
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chunyang Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
- Multimodality Preclinical Molecular Imaging Center, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
11
|
Zhou Q, Dutta D, Cao Y, Ge Z. Oxidation-Responsive PolyMOF Nanoparticles for Combination Photodynamic-Immunotherapy with Enhanced STING Activation. ACS NANO 2023; 17:9374-9387. [PMID: 37141569 DOI: 10.1021/acsnano.3c01333] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Stimulator of interferon genes (STING) activation by STING agonists has been recognized as one of the potent and promising immunotherapy strategies. However, the immunosuppressive tumor microenvironment always hinders the therapeutic efficacy of cancer immunotherapy. In this report, we present polymeric metal-organic framework (PMOF) nanoparticles (NPs) for the combination of photodynamic therapy (PDT) and enhanced STING activation to improve the immunotherapeutic efficacy. The PMOF NPs with poly(ethylene glycol) (PEG) shells were obtained via coordination between the block copolymer ligand PEG-b-PABDA consisting of 1,4-bezenedicarboxylic acid-bearing polyacrylamide (PABDA), meso-tetra(carboxyphenyl)porphyrin (TCPP), thioketal diacetic acid, and zirconyl chloride. Subsequently, the STING agonist SR-717 was loaded into the porous structure of PMOF to obtain SR@PMOF NPs which show excellent stability under the physiological conditions. After intravenous injection and tumor accumulation, light irradiation on the tumor sites results in efficient singlet oxygen (1O2) production from TCPP and cellular apoptosis to release fragmented DNA and tumor-associated antigens. Simultaneously, thioketal bonds can be broken by 1O2 to destroy the PMOF structure and rapidly release SR717. SR-717 and PDT synergistically enhance the antitumor immunity via combination photodynamic-immunotherapy due to reversal of the immunosuppressive tumor microenvironment and enhanced endogenous STING activation, which can suppress the growth of the primary and distant tumors efficiently. The oxidation-responsive SR@PMOF NPs represent a promising delivery system of STING agonists and efficient PDT NPs for simultaneous suppression of the primary and metastatic tumors via the rational combination of PDT and enhanced STING activation.
Collapse
Affiliation(s)
- Qinghao Zhou
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Debabrata Dutta
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Yufei Cao
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Zhishen Ge
- School of Chemistry, Engineering Research Center of Energy Storage Materials and Devices, Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| |
Collapse
|
12
|
Liu X, Song H, Sun T, Wang H. Responsive Microneedles as a New Platform for Precision Immunotherapy. Pharmaceutics 2023; 15:1407. [PMID: 37242649 PMCID: PMC10220742 DOI: 10.3390/pharmaceutics15051407] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/19/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Microneedles are a well-known transdermal or transdermal drug delivery system. Different from intramuscular injection, intravenous injection, etc., the microneedle delivery system provides unique characteristics for immunotherapy administration. Microneedles can deliver immunotherapeutic agents to the epidermis and dermis, where immune cells are abundant, unlike conventional vaccine systems. Furthermore, microneedle devices can be designed to respond to certain endogenous or exogenous stimuli including pH, reactive oxygen species (ROS), enzyme, light, temperature, or mechanical force, thereby allowing controlled release of active compounds in the epidermis and dermis. In this way, multifunctional or stimuli-responsive microneedles for immunotherapy could enhance the efficacy of immune responses to prevent or mitigate disease progression and lessen systemic adverse effects on healthy tissues and organs. Since microneedles are a promising drug delivery system for accurate delivery and controlled drug release, this review focuses on the progress of using reactive microneedles for immunotherapy, especially for tumors. Limitations of current microneedle system are summarized, and the controllable administration and targeting of reactive microneedle systems are examined.
Collapse
Affiliation(s)
- Xinyang Liu
- Henan Institutes of Advanced Technology, Zhengzhou University, Zhengzhou 450052, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Haohao Song
- Henan Institutes of Advanced Technology, Zhengzhou University, Zhengzhou 450052, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Tairan Sun
- The Second Affiliated Hospital of Hebei North University, Zhangjiakou 075100, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
13
|
Mukerabigwi JF, Tang R, Cao Y, Mohammed F, Zhou Q, Zhou M, Ge Z. Mitochondria-Targeting Polyprodrugs to Overcome the Drug Resistance of Cancer Cells by Self-Amplified Oxidation-Triggered Drug Release. Bioconjug Chem 2023; 34:377-391. [PMID: 36716444 DOI: 10.1021/acs.bioconjchem.2c00559] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The multi-drug resistance (MDR) of cancers is one of the main barriers for the success of diverse chemotherapeutic methods and is responsible for most cancer deaths. Developing efficient approaches to overcome MDR is still highly desirable for efficient chemotherapy of cancers. The delivery of targeted anticancer drugs that can interact with mitochondrial DNA is recognized as an effective strategy to reverse the MDR of cancers due to the relatively weak DNA-repairing capability in the mitochondria. Herein, we report on a polyprodrug that can sequentially target cancer cells and mitochondria using folic acid (FA) and tetraphenylphosphonium (TPP) targeting moieties, respectively. They were conjugated to the terminal groups of the amphiphilic block copolymer prodrugs composed of poly[oligo(ethylene glycol) methyl ether methacrylate] (POEGMA) and copolymerized monomers containing cinnamaldehyde (CNM) and doxorubicin (DOX). After self-assembly into micelles with the suitable size (∼30 nm), which were termed as TF@CNM + DOX, and upon intravenous administration, the micelles can accumulate in tumor tissues. After FA-mediated endocytosis, the endosomal acidity (∼pH 5) can trigger the release of CNM from TF@CNM + DOX micelles, followed by enhanced accumulation into the mitochondria via the TPP target. This promotes the overproduction of reactive oxygen species (ROS), which can subsequently enhance the intracellular oxidative stress and trigger ROS-responsive release of DOX into the mitochondria. TF@CNM + DOX shows great potential to inhibit the growth of DOX-resistant MCF-7 ADR tumors without observable side effects. Therefore, the tumor and mitochondria dual-targeting polyprodrug design represents an ideal strategy to treat MDR tumors through improvement of the intracellular oxidative level and ROS-responsive drug release.
Collapse
Affiliation(s)
- Jean Felix Mukerabigwi
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.,Department of Chemistry, School of Science, College of Science and Technology, University of Rwanda, Kigali, 3900 Kigali, Rwanda
| | - Rui Tang
- Neurocritical Care Unit, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Yufei Cao
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Fathelrahman Mohammed
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Qinghao Zhou
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.,CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Min Zhou
- Neurocritical Care Unit, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Zhishen Ge
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| |
Collapse
|
14
|
de Santana WMO, Pochapski DJ, Pulcinelli SH, Fontana CR, Santilli CV. Polymeric micelles–mediated photodynamic therapy. NANOMATERIALS FOR PHOTODYNAMIC THERAPY 2023:105-139. [DOI: 10.1016/b978-0-323-85595-2.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
Meng L, Ren J, Li L. Hyaluronic acid-targeted mixed micelles encapsulating hypericin for breast cancer photodynamic therapy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
16
|
Ding Y, Wang C, Ma Y, Zhu L, Lu B, Wang Y, Wang J, Dong CM, Yao Y. Tumor microenvironment responsive polypeptide-based supramolecular nanoprodrugs for combination therapy. Acta Biomater 2022. [DOI: 10.10.1016/j.actbio.2022.04.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Ding Y, Wang C, Ma Y, Zhu L, Lu B, Wang Y, Wang J, Dong CM, Yao Y. Tumor microenvironment responsive polypeptide-based supramolecular nanoprodrugs for combination therapy. Acta Biomater 2022; 146:396-405. [PMID: 35470074 DOI: 10.1016/j.actbio.2022.04.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 12/15/2022]
Abstract
Tumor microenvironment responsive nanomedicine has drawn considerable attention for combination therapy, but still remains a significant challenge for less side effects and enhanced anti-tumor efficiency. Herein, we develop a pH/ROS dual-responsive supramolecular polypeptide nanoprodrug (PFW-DOX/GOD) by using pillar[5]arene-based host-guest strategy for combined glucose degradation, chemodynamic therapy (CDT), and chemotherapy (CT). The PFW-DOX/GOD consists of a pH-responsive ferrocene/pillar[5]arene-containing polypeptide, a ROS-responsive polyprodrug, and encapsulated glucose oxidase (GOD). Upon into intracellular acidic environment, PFW-DOX/GOD exhibits rapid pH-triggered disassembly behavior. Simultaneously, the released GOD can catalyze intratumoral glucose into massive H2O2, which are further converted into highly toxic hydroxyl radicals (•OH) by the catalysis of ferrocene via the Fenton reaction. Thereafter, induced by the ROS-responsive cleavage of thioketal linkage, the conjugated DOX prodrug was released and activated. The combined glucose degradation, chemodynamic therapy (CDT), and chemotherapy (CT) of PFW-DOX/GOD present anti-tumor effect with 96% of tumor inhibitory rate (TIR). Therefore, such tumor microenvironment-responsive supramolecular polypeptide nanoprodrugs represent a potential candidate for combination therapy with minimal side effects. STATEMENT OF SIGNIFICANCE: In this work, a tumor microenvironment-responsive supramolecular polypeptide nanoprodrug (PFW-DOX/GOD) was prepared via pillar[5]arene-based host-guest interactions, and presented low side effects and high tumor accumulation owing to the diameters of about 200 nm and surface PEG segment. After pH-responsive release of GOD in the intracellular acidic environment, the cascade catalytic reactions including GOD-catalyzed degradation of intratumoral glucose and Fenton reaction, effectively happened to generate •OH for chemodynamic therapy (CDT), which subsequently induced the cleavage of thioketal linkage to activate free DOX for chemotherapy (CT). Collectively, this supramolecular polypeptide nanoprodrugs provide a promising strategy for combination therapy with synergetic anti-tumor effect.
Collapse
Affiliation(s)
- Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China.
| | - Chenwei Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Yuxuan Ma
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Lvming Zhu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Bing Lu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Yang Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Jin Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China
| | - Chang-Ming Dong
- Joint Research Center for Precision Medicine, School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, PR China.
| |
Collapse
|
18
|
Itoo AM, Paul M, Ghosh B, Biswas S. Oxaliplatin delivery via chitosan/vitamin E conjugate micelles for improved efficacy and MDR-reversal in breast cancer. Carbohydr Polym 2022; 282:119108. [PMID: 35123744 DOI: 10.1016/j.carbpol.2022.119108] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 12/18/2022]
Abstract
A bioinspired chitosan/vitamin E conjugate (Ch/VES, 1:4) was synthesized, optimized based on chitosan's molecular weight (15, 300 kDa), and was assembled to entrap oxaliplatin (OXPt). 1H NMR, infrared spectroscopy, chromatography, X-ray photoelectron spectroscopy, X-ray diffraction, drug release, hemolysis, and stability studies were performed to characterize OXPt@Ch/VES micelles. The therapeutic efficacy of the micelles was tested in vitro in ER+/PR+/HER2- and triple-negative sensitive/resistant breast cancer cells, MCF-7 and MDA-MB-231 via cellular uptake, cytotoxicity, nuclear staining, DNA fragmentation, mitochondrial membrane potential, ROS generation, apoptosis, and cell cycle assays and in vivo using 4T1(Luc)-tumor-bearing mice. OXPt@Ch/VES Ms exhibited decreased IC50 towards MCF-7, MDA-MB-231 (sensitive/resistant) than OXPt. OXPt@Ch/VES Ms caused extensive DNA damage, mitochondrial depolarization, apoptosis, and cell-growth arrest (G2/M). OXPt@Ch/VES Ms treatment retarded tumor growth significantly, prolonged survival, and decreased nephrotoxicity than OXPt. The OXPt@Ch/VES Ms could serve as a potential nanomedicine to overcome conventional OXPt-mediated drug resistance/nephrotoxicity in breast cancer.
Collapse
Affiliation(s)
- Asif Mohd Itoo
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Milan Paul
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Balaram Ghosh
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India.
| |
Collapse
|
19
|
Applications of the ROS-Responsive Thioketal Linker for the Production of Smart Nanomedicines. Polymers (Basel) 2022; 14:polym14040687. [PMID: 35215600 PMCID: PMC8874672 DOI: 10.3390/polym14040687] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 12/16/2022] Open
Abstract
Reactive oxygen species (ROS)-sensitive drug delivery systems (DDS) specifically responding to altered levels of ROS in the pathological microenvironment have emerged as an effective means to enhance the pharmaceutical efficacy of conventional nanomedicines, while simultaneously reducing side effects. In particular, the use of the biocompatible, biodegradable, and non-toxic ROS-responsive thioketal (TK) functional group in the design of smart DDS has grown exponentially in recent years. In the design of TK-based DDS, different technological uses of TK have been proposed to overcome the major limitations of conventional DDS counterparts including uncontrolled drug release and off-target effects. This review will focus on the different technological uses of TK-based biomaterials in smart nanomedicines by using it as a linker to connect a drug on the surface of nanoparticles, form prodrugs, as a core component of the DDS to directly control its structure, to control the opening of drug-releasing gates or to change the conformation of the nano-systems. A comprehensive view of the various uses of TK may allow researchers to exploit this reactive linker more consciously while designing nanomedicines to be more effective with improved disease-targeting ability, providing novel therapeutic opportunities in the treatment of many diseases.
Collapse
|
20
|
Yates-Alston S, Sarkar S, Cochran M, Kuthirummal N, Levi N. Hybrid donor-acceptor polymer nanoparticles and combination antibiotic for mitigation of pathogenic bacteria and biofilms. J Microbiol Methods 2021; 190:106328. [PMID: 34536464 DOI: 10.1016/j.mimet.2021.106328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 11/19/2022]
Abstract
Biofilms pose a significant clinical problem in skin and soft tissue infections. Their resistance to antibiotics has spurred investigations into alternative treatments, such as nanoparticle-mediated photothermal ablation. Non-toxic Hybrid Donor- Acceptor (DA) Polymer nanoParticles (H-DAPPs) were developed for fluorescence imaging (using poly(3-hexylthiophene-2,5 diyl) (P3HT)) and rapid, near-infrared photothermal ablation (NIR- PTA) (using poly[4,4-bis(2-ethylhexyl)-cyclopenta[2,1-b;3,4-b']dithiophene-2,6-diyl-alt-2,1,3-benzoselenadiazole-4,7-diyl] (PCPDTBSe)). H-DAPPs were evaluated alone, and in combination with antibiotics, against planktonic S. aureus and S. pyogenes, and S. aureus biofilms. H-DAPPs NIR-PTA (15-700 μg/ mL) can generate rapid temperature changes of 27.6-73.1 °C, which can eradicate planktonic bacterial populations and reduce biofilm bacterial viability by more than 4- log (> 99.99%) with exposure to 60 s of 800 nm light. Reductions were confirmed via confocal analysis, which suggested that H-DAPPs PTA caused bacterial inactivation within the biofilms, but did not significantly reduce biofilm polysaccharides. SEM imaging revealed structural changes in biofilms after H-DAPPs PTA. S. aureus biofilms challenged with 100 μg/mL of H-DAPPs (H-DAPPs-100) to induce an average temperature of 55.1 °C, and the minimum biofilm eradication concentration (MBEC) of clindamycin, resulted in up to ~3- log decrease in bacterial viability compared to untreated biofilms and those administered H-DAPPs-100 PTA only, and up to ~2- log compared to biofilms administered only clindamycin. This study demonstrates that polymer nanoparticle PTA can mitigate biofilm infection and may improve antimicrobial efficacy.
Collapse
Affiliation(s)
- Shaina Yates-Alston
- Department of Plastic and Reconstructive Surgery, Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA
| | - Santu Sarkar
- Department of Plastic and Reconstructive Surgery, Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA
| | - Matthew Cochran
- Department of Plastic and Reconstructive Surgery, Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA
| | | | - Nicole Levi
- Department of Plastic and Reconstructive Surgery, Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA.
| |
Collapse
|
21
|
Deng K, Yu H, Li JM, Li KH, Zhao HY, Ke M, Huang SW. Dual-step irradiation strategy to sequentially destroy singlet oxygen-responsive polymeric micelles and boost photodynamic cancer therapy. Biomaterials 2021; 275:120959. [PMID: 34147717 DOI: 10.1016/j.biomaterials.2021.120959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022]
Abstract
Nanotechnology provides a powerful tool to overcome many disadvantages of small-molecule photosensitizers for photodynamic cancer therapy, such as hydrophobicity, rapid blood clearance, low accumulation in tumor tissue and low cell penetration, etc. The occurrence of quench in photosensitizer-loaded nanoparticle greatly downregulates the ability to generate singlet oxygen with light irradiation. Stimuli-responsive nanocarriers can improve the efficacy of PDT to a certain extent. However, insufficient release of photosensitizer from either endogenous or exogenous stimuli responsive nanocarriers in the short period of light irradiation restricts full usage of the photosensitizer delivered into cancer cells. We here report a dual-step light irradiation strategy to enhance the efficacy of cancer PDT. Ce6 as a photosensitizer is loaded in singlet oxygen-sensitive micelles (Ce6-M) via self-assembly of amphiphilic polymer mPEG2000-TK-C16. After co-incubation of Ce6-M with cancer cells or i.v. injection of Ce6-M, cancer cells or tumor tissues are irradiated with light for a short time to trigger Ce6 release, and 2 h later, re-irradiated for relatively long time. The sufficient release of Ce6 in the period between twice light irradiation significantly improves the generation of singlet oxygen, leading to more efficient cancer therapeutic effects of dual-step irradiation than that of single-step irradiation for the same total irradiation time.
Collapse
Affiliation(s)
- Kai Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China; Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China.
| | - Hui Yu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Jia-Mi Li
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Kun-Heng Li
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Hong-Yang Zhao
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Min Ke
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Shi-Wen Huang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
22
|
Yang DC, Wang S, Weng XL, Zhang HX, Liu JY, Lin Z. Singlet Oxygen-Responsive Polymeric Nanomedicine for Light-Controlled Drug Release and Image-Guided Photodynamic-Chemo Combination Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:33905-33914. [PMID: 34278780 DOI: 10.1021/acsami.1c09044] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Coencapsulation of chemotherapeutic agents and photosensitizers into nanocarriers can help to achieve a combination of chemotherapy and photodynamic therapy for superior antitumor effects. However, precise on-demand drug release remains a major challenge. In addition, the loaded photosensitizers usually tend to aggregate, which can significantly weaken their fluorescent signals and photodynamic activities. To address these issues, herein, a smart nanocarrier termed as singlet oxygen-responsive nanoparticle (SOR-NP) was constructed by introducing singlet oxygen (1O2)-sensitive aminoacrylate linkers into amphiphilic mPEG-b-PCL copolymers. Boron dipyrromethene (BDP) and paclitaxel (PTX) as model therapeutic agents were coloaded into an 1O2-responsive nanocarrier for realizing light-controlled drug release and combination cancer treatment. This polymeric nanocarrier could substantially relieve the aggregation of encapsulated BDP due to the presence of a long hydrophobic chain. Therefore, the formed SOR-NPBDP/PTX nanodrug could generate bright fluorescent signals and high levels of 1O2, which could mediate cell death via PDT and rupture aminoacrylate linker simultaneously, leading to collapse of SOR-NPBDP/PTX and subsequent PTX release. The light-triggered drug release and combined anticancer effects of SOR-NPBDP/PTX were validated in HepG2 and MCF-7 cancer cells and H22 tumor-bearing mice. This study provides a promising strategy for tumor-specific drug release and selective photodynamic-chemo combination treatment.
Collapse
Affiliation(s)
- De-Chao Yang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Shuai Wang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Xiao-Lu Weng
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Hong-Xia Zhang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Jian-Yong Liu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou 350108, China
- State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Zhonghui Lin
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
23
|
Liu J, Ye LY, Xiong WH, Liu T, Yang H, Lei J. A cerium oxide@metal-organic framework nanoenzyme as a tandem catalyst for enhanced photodynamic therapy. Chem Commun (Camb) 2021; 57:2820-2823. [PMID: 33605969 DOI: 10.1039/d1cc00001b] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
An O2 self-evolving core-shell theranostic nanohybrid was developed by encapsulating a nanoenzyme cerium oxide (CeOx) in a metal-organic framework (MOF). The hybrid reveals a 9-fold higher apoptotic percentage than bare CeOx in a harsh hypoxic microenvironment through tandem homogenous catalysis. Simultaneously, the oxygen-promoted therapeutic efficiency was self-monitored by the hybrid with caspase-3 activation, paving the way for MOF-functionalized nanoenzymes in theranostics.
Collapse
Affiliation(s)
- Jintong Liu
- Department of Chemistry, College of Sciences, Nanjing Agricultural University, Nanjing 210095, P. R. China. and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Lin Yao Ye
- Department of Chemistry, College of Sciences, Nanjing Agricultural University, Nanjing 210095, P. R. China.
| | - Wei Hong Xiong
- Department of Chemistry, College of Sciences, Nanjing Agricultural University, Nanjing 210095, P. R. China.
| | - Tianrui Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Hong Yang
- Department of Chemistry, College of Sciences, Nanjing Agricultural University, Nanjing 210095, P. R. China.
| | - Jianping Lei
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| |
Collapse
|
24
|
Bobde Y, Paul M, Patel T, Biswas S, Ghosh B. Polymeric micelles of a copolymer composed of all-trans retinoic acid, methoxy-poly(ethylene glycol), and b-poly(N-(2 hydroxypropyl) methacrylamide) as a doxorubicin-delivery platform and for combination chemotherapy in breast cancer. Int J Pharm 2021; 606:120866. [PMID: 34237409 DOI: 10.1016/j.ijpharm.2021.120866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/20/2021] [Accepted: 07/02/2021] [Indexed: 12/26/2022]
Abstract
Delivery of combination chemotherapeutic agents to the tumor via nanovesicles has the potential for superior tumor suppression and reduced toxicity. Herein, we prepare a block copolymer (mPH-RA) composed of methoxy-poly(ethylene glycol) (mPEG), b-poly(N-(2 hydroxypropyl) methacrylamide) (pHPMA), and all-trans retinoic acid (ATRA) by conjugating ATRA to the pre-formed copolymer, mPEG-b-pHPMA(mP-b-pH). Doxorubicin-loaded micelles, Dox@mP-b-pH, and Dox@mPH-RA were characterized by determining particle size, zeta potential, % DL, EE, Dox release, hemolysis study, and by DSC. The Dox@mPH-RA micelles (mPH-RA: Dox ratios of 10:0.5-2) displayed nano-size (36-45 nm), EE. 26-74%, and DL. 2.9-5.6%. Dox@mPH-RA micelles displayed the highest penetrability and cytotoxicity than free Dox and Dox@mP-b-pH micelles in breast cancer cell lines. Dox@mPH-RA exhibited the highest induction of apoptosis (94.1 ± 3%) than Dox (52.1 ± 4.5%), and Dox@mP-b-pH (81.7 ± 3%), and arrested cells in the highest population in G2 and S phase. Dox@mPH-RA increased the t1/2 and Cmax of Dox and demonstrated improved therapeutic efficacy and highest Dox distribution to the tumor. The Dox@mPH-RA increased the levels of apoptosis markers, caspase 3, 7, Ki-67, and caused the highest DNA fragmentation. The presence of RA improved the micelles' physicochemical properties, Dox-loading ability, and the therapeutic potential in Dox@mPH-RA via the combination therapeutic strategy.
Collapse
Affiliation(s)
- Yamini Bobde
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science - Pilani, Hyderabad Campus, Medchal, Hyderabad 500078, India; Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science - Pilani, Hyderabad Campus, Medchal, Hyderabad 500078, India
| | - Milan Paul
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science - Pilani, Hyderabad Campus, Medchal, Hyderabad 500078, India
| | - Tarun Patel
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science - Pilani, Hyderabad Campus, Medchal, Hyderabad 500078, India
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science - Pilani, Hyderabad Campus, Medchal, Hyderabad 500078, India.
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science - Pilani, Hyderabad Campus, Medchal, Hyderabad 500078, India.
| |
Collapse
|
25
|
Peng F, Chen Y, Liu J, Xing Z, Fan J, Zhang W, Qiu F. Facile design of gemini surfactant-like peptide for hydrophobic drug delivery and antimicrobial activity. J Colloid Interface Sci 2021; 591:314-325. [PMID: 33621783 DOI: 10.1016/j.jcis.2021.02.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 02/08/2023]
Abstract
Recently, many kinds of gemini-type amphiphilic peptides have been designed and shown their advantage as self-assembling nanomaterials. In this study, we proposed a simple strategy to design gemini surfactant-like peptides, which are only composed of natural amino acids and can be easily obtained by conventional peptide sythnesis. Taking two prolines as the turn-forming units, a peptide named APK was designed. The petide has a linear sequence but naturally takes the conformation like a gemini surfactant. Compared with a single-tailed surfactant-like peptide A6K, APK showed much stronger ability to undergo self-assembly and to encapsulate hydrophobic pyrene. Several hydrophobic drugs including paclitaxel, doxorubicin, etomidate and propofol were encapsulated by APK, and the corresponding formulations showed anti-tumor or anesthetic efficacy comparable to their respective clinical formulations. Furthermore, APK could inhibit the growth of different microorganisms including E. coli, S. aureus and C. albicans. Etomidate and propofol formulations encapsulated by APK also showed strong antimicrobial activity. Taking APK as an example, our study indicated a straightforward strategy to design gemini surfactant-like peptides, which could be potential nanomaterials for exploring hydrophobic drug formulations with efficacy, safety and self-antimicrobial activity.
Collapse
Affiliation(s)
- Fei Peng
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongzhu Chen
- Periodical Press of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Liu
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhihua Xing
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Fan
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wensheng Zhang
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Feng Qiu
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China; National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
26
|
Li G, Zhao M, Zhao L. Lysine-mediated hydroxyethyl starch-10-hydroxy camptothecin micelles for the treatment of liver cancer. Drug Deliv 2021; 27:519-529. [PMID: 32228107 PMCID: PMC7170360 DOI: 10.1080/10717544.2020.1745329] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Liver cancer is a malignant tumor with extremely high morbidity and mortality. At present, traditional chemotherapy is still the most commonly used therapeutic approach. However, serious side effects lead to the treatment of liver cancer is not ideal. Therefore, it is imperative to develop a new drug delivery system based on nanotechnology and liver cancer microenvironment. In this study, a pH/reduction/α-amylase multi-sensitive hydroxyethyl starch-10-hydroxy camptothecin micelles (HES-10-HCPT-SS-Ly) targeting over-expressed amino acid (AA) transporters on the surface of liver cancer cell by applying lysine were successfully synthesized. The prepared micelles showed regular structure, suitable particle size, and intelligent drug release property. Compared with conventional HES-10-HCPT micelles and 10-HCPT injection, HES-10-HCPT-SS-Ly micelles demonstrated better in vitro anti-proliferative capability toward human liver cancer Hep-G2 cells and greater antitumor efficiency against nude mouse with Hep-G2 tumor. These findings suggest that HES-10-HCPT-SS-Ly micelles may be a promising nanomedicine for treatment of liver cancer.
Collapse
Affiliation(s)
- Guofei Li
- Department of Pharmacy, Shengjing Hospital, China Medical University, Shenyang, China
| | - Mingming Zhao
- Department of Pharmacy, Shengjing Hospital, China Medical University, Shenyang, China
| | - Limei Zhao
- Department of Pharmacy, Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
27
|
Wyers D, Goris T, De Smet Y, Junkers T. Amino acid acrylamide mimics: creation of a consistent monomer library and characterization of their polymerization behaviour. Polym Chem 2021. [DOI: 10.1039/d1py00735a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel consistent approach to mimic the structure of biopolymers via precision polymer synthesis with reversible deactivation radical polymerization (RDRP).
Collapse
Affiliation(s)
- Dries Wyers
- Polymer Reaction Design Group, School of Chemistry, Monash University, 19 Rainforest Walk, Clayton, VIC 3800, Australia
| | - Toon Goris
- Polymer Reaction Design Group, School of Chemistry, Monash University, 19 Rainforest Walk, Clayton, VIC 3800, Australia
| | - Yana De Smet
- Polymer Reaction Design Group, School of Chemistry, Monash University, 19 Rainforest Walk, Clayton, VIC 3800, Australia
| | - Tanja Junkers
- Polymer Reaction Design Group, School of Chemistry, Monash University, 19 Rainforest Walk, Clayton, VIC 3800, Australia
| |
Collapse
|
28
|
Liu G, Lovell JF, Zhang L, Zhang Y. Stimulus-Responsive Nanomedicines for Disease Diagnosis and Treatment. Int J Mol Sci 2020; 21:E6380. [PMID: 32887466 PMCID: PMC7504550 DOI: 10.3390/ijms21176380] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
Stimulus-responsive drug delivery systems generally aim to release the active pharmaceutical ingredient (API) in response to specific conditions and have recently been explored for disease treatments. These approaches can also be extended to molecular imaging to report on disease diagnosis and management. The stimuli used for activation are based on differences between the environment of the diseased or targeted sites, and normal tissues. Endogenous stimuli include pH, redox reactions, enzymatic activity, temperature and others. Exogenous site-specific stimuli include the use of magnetic fields, light, ultrasound and others. These endogenous or exogenous stimuli lead to structural changes or cleavage of the cargo carrier, leading to release of the API. A wide variety of stimulus-responsive systems have been developed-responsive to both a single stimulus or multiple stimuli-and represent a theranostic tool for disease treatment. In this review, stimuli commonly used in the development of theranostic nanoplatforms are enumerated. An emphasis on chemical structure and property relationships is provided, aiming to focus on insights for the design of stimulus-responsive delivery systems. Several examples of theranostic applications of these stimulus-responsive nanomedicines are discussed.
Collapse
Affiliation(s)
- Gengqi Liu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China;
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, The State University of New York at Buffalo, Buffalo, NY 14260, USA;
| | - Lei Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China;
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China;
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| |
Collapse
|
29
|
Yang H, Tong Z, Sun S, Mao Z. Enhancement of tumour penetration by nanomedicines through strategies based on transport processes and barriers. J Control Release 2020; 328:28-44. [PMID: 32858072 DOI: 10.1016/j.jconrel.2020.08.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Nanomedicines for antitumour therapy have been widely studied in recent decades, but only a few have been used in clinical applications. One of the most important reasons is the poor tumour permeability of the nanomedicines. In this three-part review, intravascular, transvascular and extravascular transport were introduced one by one according to their roles in the overall process of nanomedicine transport into tumours. Transportation obstacles, such as elevated interstitial fluid pressure (IFP), abnormal blood vessels, dense tumour extracellular matrix (ECM) and binding site barriers (BSB), were each discussed in the context of the respective transport processes. Furthermore, homologous resolution strategies were summarized on the basis of each transportation obstacle, such as the normalization of blood vessels, regulation of the tumour microenvironment (TME) and application of transformable nanoparticles. At the end of this review, we propose holistic, concrete, and innovative views for better tumour penetration of nanomedicines.
Collapse
Affiliation(s)
- Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Zongrui Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Shichao Sun
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| |
Collapse
|
30
|
Wang D, Wang S, Xia Y, Liu S, Jia R, Xu G, Zhan J, Lu Y. Preparation of ROS-responsive core crosslinked polycarbonate micelles with thioketal linkage. Colloids Surf B Biointerfaces 2020; 195:111276. [PMID: 32763765 DOI: 10.1016/j.colsurfb.2020.111276] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 11/17/2022]
Abstract
Herein, we prepared novel reactive oxygen species (ROS) responsive core crosslinked (CCL/TK) polycarbonate micelles conveniently by click reaction between amphiphilic diblock copolymer poly(ethylene glycol)-poly(5-methyl-5-propargylxycar-bonyl-1,3-dioxane-2-one) (PEG-PMPC) with pendant alkynyl group and thioketal containing azide derivative bis (2-azidoethyl) 3, 3'- (propane-2, 2-diylbis (sulfanediyl)) dipropanoate (TK-N3). The CCL/TK micelles were obtained with small size of 146.4 nm, showing excellent stability against dilution and high doxorubicin (DOX) loading. In vitro toxicity tests demonstrated that the obtained CCL/TK micelles have good biocompatibility and low toxicity with cell viability above 95 %. Furthermore, DOX-loaded CCL/TK micelles showed significantly superior toxicity with IC50 values for HeLa and MCF-7 cells about 3.74 μg/mL and 3.91 μg/mL, respectively. Confocal laser scanning microscope (CLSM) and flow cytometry showed excellent internalization efficiency and intracellular drug release of DOX-loaded CCL/TK micelles. The obtained ROS-responsive CCL/TK micelles showed great potential for anticancer drug delivery.
Collapse
Affiliation(s)
- Deqi Wang
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Song Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, PR China
| | - Yingchun Xia
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Simeng Liu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Ruixin Jia
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Gege Xu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Junjie Zhan
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Yanbing Lu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China; State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, PR China.
| |
Collapse
|
31
|
Stimuli-responsive nano-assemblies for remotely controlled drug delivery. J Control Release 2020; 322:566-592. [DOI: 10.1016/j.jconrel.2020.03.051] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 12/30/2022]
|
32
|
Wang M, Yang Q, Li M, Zou H, Wang Z, Ran H, Zheng Y, Jian J, Zhou Y, Luo Y, Ran Y, Jiang S, Zhou X. Multifunctional Nanoparticles for Multimodal Imaging-Guided Low-Intensity Focused Ultrasound/Immunosynergistic Retinoblastoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:5642-5657. [PMID: 31940169 DOI: 10.1021/acsami.9b22072] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Retinoblastoma (RB) is prone to delayed diagnosis or treatment and has an increased likelihood of metastasizing. Thus, it is crucial to perform an effective imaging examination and provide optimal treatment of RB to prevent metastasis. Nanoparticles that support diagnostic imaging and targeted therapy are expected to noninvasively integrate tumor diagnosis and treatment. Herein, we report a multifunctional nanoparticle for multimodal imaging-guided low-intensity focused ultrasound (LIFU)/immunosynergistic RB therapy. Magnetic hollow mesoporous gold nanocages (AuNCs) conjugated with Fe3O4 nanoparticles (AuNCs-Fe3O4) were prepared to encapsulate muramyl dipeptide (MDP) and perfluoropentane (PFP). The multimodal imaging capabilities, antitumor effects, and dendritic cell (DC) activation capacity of these nanoparticles combined with LIFU were explored in vitro and in vivo. The biosafety of AuNCs-Fe3O4/MDP/PFP was also evaluated systematically. The multifunctional magnetic nanoparticles enhanced photoacoustic (PA), ultrasound (US), and magnetic resonance (MR) imaging in vivo and in vitro, which was helpful for diagnosis and efficacy evaluation. Upon accumulation in tumors via a magnetic field, the nanoparticles underwent phase transition under LIFU irradiation and MDP was released. A combined effect of AuNCs-Fe3O4/MDP/PFP and LIFU was recorded and verified. AuNCs-Fe3O4/MDP/PFP enhanced the therapeutic effect of LIFU and led to direct apoptosis/necrosis of tumors, while MDP promoted DC maturation and activation and activated the ability of DCs to recognize and clear tumor cells. By enhancing PA/US/MR imaging and inhibiting tumor growth, the multifunctional AuNC-Fe3O4/MDP/PFP nanoparticles show great potential for multimodal imaging-guided LIFU/immunosynergistic therapy of RB. The proposed nanoplatform facilitates cancer theranostics with high biosafety.
Collapse
Affiliation(s)
- Menglei Wang
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Qiming Yang
- Department of Orthopedic , The First Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Meng Li
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Hongmi Zou
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yuanyi Zheng
- Shanghai Institute of Ultrasound in Medicine , Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233 , P. R. China
| | - Jia Jian
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yu Zhou
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yindeng Luo
- Department of Radiology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yijun Ran
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Shaoqiu Jiang
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Xiyuan Zhou
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| |
Collapse
|