1
|
Ghosh R, Kumar M, Kumar S, Komal K, Sharma R, Kurmi BD. Small molecule therapeutics for receptor-mediated targeting through liposomes in breast cancer treatment: A comprehensive review. Bioorg Chem 2025; 160:108442. [PMID: 40199009 DOI: 10.1016/j.bioorg.2025.108442] [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: 01/24/2025] [Revised: 03/06/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Breast cancer (BC) remains a significant global health challenge, with conventional treatment approaches such as surgery, chemotherapy, and radiation therapy. These approaches face limitations in targeting, toxicity, and efficacy. Liposomal drug delivery systems have emerged as promising tools for targeted breast cancer therapies. Liposomes can encapsulate both hydrophilic and hydrophobic drugs, improve drug distribution, and reduce the side effects. Passive targeting exploits the enhanced permeability and retention effect in tumor tissues, whereas active targeting employs small molecule ligands such as aptamers, folic acid (FA), transferrin, and monoclonal antibodies to specifically bind to overexpressed receptors on cancer cells. Aptamer-functionalized liposomes exhibit high specificity and affinity, folate and transferrin receptor targeting enhances cellular uptake and cytotoxicity, and antibody-conjugated liposomes improve drug delivery and efficacy by targeting specific antigens. Dual-responsive liposomes are sensitive to multiple stimuli and further enhance targeting precision. However, challenges remain, including tumor heterogeneity, limited penetration, and potential immunogenicity. Current research has focused on developing stable and effective formulations and exploring combination-targeting strategies to overcome these limitations. With further advancements, targeted liposomal drug delivery systems hold great promise in improving breast cancer treatment outcomes and reducing adverse effects.
Collapse
Affiliation(s)
- Rashmi Ghosh
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India.
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Rohit Sharma
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga 142001, Punjab, India
| |
Collapse
|
2
|
Han X, Zhang X, Kang L, Feng S, Li Y, Zhao G. Peptide-modified nanoparticles for doxorubicin delivery: Strategies to overcome chemoresistance and perspectives on carbohydrate polymers. Int J Biol Macromol 2025; 299:140143. [PMID: 39855525 DOI: 10.1016/j.ijbiomac.2025.140143] [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: 12/16/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Chemotherapy serves as the primary treatment for cancers, facing challenges due to the emergence of drug resistance. Combination therapy has been developed to combat cancer drug resistance, yet it still suffers from lack of specific targeting of cancer cells and poor accumulation at the tumor site. Consequently, targeted administration of chemotherapy medications has been employed in cancer treatment. Doxorubicin (DOX) is one of the most frequently used chemotherapeutics, functioning by inhibiting topoisomerase activity. Enhancing the anti-cancer effects of DOX and overcoming drug resistance can be accomplished via delivery by nanoparticles. This review will focus on the development of peptide-DOX conjugates, the functionalization of nanoparticles with peptides, the co-delivery of DOX and peptides, as well as the theranostic use of peptide-modified nanoparticles in cancer treatment. The peptide-DOX conjugates have been designed to enhance the targeted delivery to cancer cells by interacting with receptors that are overexpressed on tumor surfaces. Moreover, nanoparticles can be modified with peptides to improve their uptake in tumor cells via endocytosis. Nanoparticles have the ability to co-deliver DOX along with therapeutic peptides for enhanced cancer treatment. Finally, nanoparticles modified with peptides can offer theranostic capabilities by facilitating both imaging and the delivery of DOX (chemotherapy).
Collapse
Affiliation(s)
- Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xue Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, China
| | - Longdan Kang
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Shuai Feng
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Ge Zhao
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Mamidi N, Franco De Silva F, Orash Mahmoudsalehi A. Advanced disease therapeutics using engineered living drug delivery systems. NANOSCALE 2025; 17:7673-7696. [PMID: 40040419 DOI: 10.1039/d4nr05298f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Biological barriers significantly impede the delivery of nanotherapeutics to diseased tissues, diminishing therapeutic efficacy across pathologies such as cancer and inflammatory disorders. Although conventional strategies integrate multifunctional designs and molecular components into nanomaterials (NMs), many approaches remain insufficient to overcome these barriers. Key challenges, including inadequate drug accumulation at target sites and nonspecific biodistribution, persist in nanotherapeutic development. NMs, which harness the ability to precisely modulate drug delivery spatiotemporally and control release kinetics, represent a transformative platform for targeted cancer therapy. In this review, we highlight the biological obstacles limiting effective cancer treatment and evaluate how stimuli-responsive NMs address these constraints. By leveraging exogenous and endogenous stimuli, such NMs improve therapeutic specificity, reduce off-target effects, and amplify drug activity within pathological microenvironments. We systematically analyze the rational design and synthesis of stimuli-responsive NMs, driven by advances in oncology, biomaterials science, and nanoscale engineering. Furthermore, we highlight advances across NM classes-including polymeric, lipid-based, inorganic, and hybrid systems and explore functionalization approaches using targeting ligands, antibodies, and biomimetic coatings. Diverse delivery strategies are evaluated, such as small-molecule prodrug activation, peptide- and protein-based targeting, nucleic acid payloads, and engineered cell-mediated transport. Despite the promise of stimuli-responsive NMs, challenges such as biocompatibility, scalable fabrication, and clinical translation barriers must be addressed. By elucidating structure-function relationships and refining stimulus-triggered mechanisms, these NMs pave the way for transformative precision oncology strategies, enabling patient-specific therapies with enhanced efficacy and safety. This synthesis of interdisciplinary insights aims to catalyze innovation in next-generation nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Wisconsin Center for Nanobiosystems, School of Pharmacy, University of Wisconsin-Madison, Wisconsin-53705, USA.
| | - Fátima Franco De Silva
- Department of Food Engineering, Tecnologico de Monterrey, Monterrey, Nuevo Leon-64849, Mexico
| | - Amin Orash Mahmoudsalehi
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Av. Eugenio Garza Sada 2501 Sur, Monterrey, Nuevo Leon-64849, Mexico
| |
Collapse
|
4
|
Zhang J, Yang X, Chang Z, Zhu W, Ma Y, He H. Polymeric nanocarriers for therapeutic gene delivery. Asian J Pharm Sci 2025; 20:101015. [PMID: 39931356 PMCID: PMC11808530 DOI: 10.1016/j.ajps.2025.101015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 02/13/2025] Open
Abstract
The recent commercialization of gene products has sparked significant interest in gene therapy, necessitating efficient and precise gene delivery via various vectors. Currently, viral vectors and lipid-based nanocarriers are the predominant choices and have been extensively investigated and reviewed. Beyond these vectors, polymeric nanocarriers also hold the promise in therapeutic gene delivery owing to their versatile functionalities, such as improving the stability, cellar uptake and endosomal escape of nucleic acid drugs, along with precise delivery to targeted tissues. This review presents a brief overview of the status quo of the emerging polymeric nanocarriers for therapeutic gene delivery, focusing on key cationic polymers, nanocarrier types, and preparation methods. It also highlights targeted diseases, strategies to improve delivery efficiency, and potential future directions in this research area. The review is hoped to inspire the development, optimization, and clinical translation of highly efficient polymeric nanocarriers for therapeutic gene delivery.
Collapse
Affiliation(s)
- Jiayuan Zhang
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, School of Pharmacy, Qinghai Minzu University, Xining 810007, China
| | - Xinyu Yang
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zhichao Chang
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wenwei Zhu
- Hepatobiliary Surgery, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuhua Ma
- Key Laboratory for Tibet Plateau Phytochemistry of Qinghai Province, School of Pharmacy, Qinghai Minzu University, Xining 810007, China
| | - Haisheng He
- Key Laboratory of Smart Drug Delivery of Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
5
|
Mojarad-Jabali S, Roh KH. Peptide-based inhibitors and nanoparticles: Emerging therapeutics for Alzheimer's disease. Int J Pharm 2025; 669:125055. [PMID: 39653296 DOI: 10.1016/j.ijpharm.2024.125055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/25/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease (AD) is an age-related progressive neurodegenerative disorder characterized by memory loss, cognitive decline, and behavioral changes, impacting millions of individuals worldwide. Despite significant research into its cellular and molecular mechanisms, no cure has been found to treat AD to date. For over two decades, research aimed at treating AD has focused on targeting amyloid-β (Aβ); however, these strategies have not demonstrated substantial effectiveness. Consequently, research is now expanding towards targeting other hallmarks of the disease, such as tau protein and brain metal ions. Among potential therapeutics against these pathophysiological targets, peptide-based inhibitors are notable for their high selectivity and low toxicity. Despite these advantages, they face obstacles such as a short half-life in vivo and low efficiencies in crossing the blood-brain barrier (BBB). The use of nanoparticles (NPs) to deliver peptide-based inhibitors to the brain offers unique advantages, such as enhanced stability against degradation, improvement in targeted delivery, and reduced potential for immunogenic responses. This review aims to provide a comprehensive overview of emerging peptides tested as treatments for AD against Aβ, tau protein, and brain metal ions and to evaluate NPs as a means to overcome the limitations. These peptide-based inhibitors are promising, as they not only alleviate symptoms but also aim to prevent progressive neuronal loss, and NPs can be highly effective in delivering these inhibitors.
Collapse
Affiliation(s)
- Solmaz Mojarad-Jabali
- Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kyung-Ho Roh
- Department of Chemical and Materials Engineering, University of Alabama in Huntsville, Huntsville, AL 35899, United States; Biotechnology Science and Engineering Program, University of Alabama in Huntsville, Huntsville, AL 35899, United States.
| |
Collapse
|
6
|
Pani I, Hardt M, Glikman D, Braunschweig B. Photo-induced drug release at interfaces with arylazopyrazoles. Chem Sci 2024:d4sc04837g. [PMID: 39464617 PMCID: PMC11503751 DOI: 10.1039/d4sc04837g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
Smart responsive materials have spurred the progress in high-precision drug delivery. Enormous attention has been given to characterizing drug release in bulk aqueous solutions, however, aqueous-hydrophobic interfaces are vital components of biological systems which serve as the point of entry into cells. These interfaces are involved in many key biomolecular interactions, and while the potential for drug molecules to adsorb to these interfaces is recognized, their specific role in the context of drug release remains largely unexplored. We present a fundamental investigation on the release of encapsulated drugs at the air-water interface as a representative model to mimic the organic/aqueous interface of cells. Combining the advantages of light as an external stimulus and the superiority of arylazopyrazoles (AAP) over conventional azobenzene photoswitches, we report a micellar nanocarrier for the capture and release of the chemotherapeutic drug doxorubicin. Using a powerful combination of interface-sensitive techniques such as the Langmuir-Blodgett technique, surface tensiometry, and the interface-specific vibrational sum-frequency generation spectroscopy, we demonstrate the photoresponsive release of doxorubicin encapsulated in the micelles of AAP photosurfactants to the air-water interface. Complementary fluorescence measurements corroborate additional drug release in bulk aqueous solutions.
Collapse
Affiliation(s)
- Ipsita Pani
- Institute of Physical Chemistry, Center for Soft Nanoscience (SoN), University of Münster Corrensstraße 28-30 Münster 48149 Germany
| | - Michael Hardt
- Institute of Physical Chemistry, Center for Soft Nanoscience (SoN), University of Münster Corrensstraße 28-30 Münster 48149 Germany
| | - Dana Glikman
- Institute of Physical Chemistry, Center for Soft Nanoscience (SoN), University of Münster Corrensstraße 28-30 Münster 48149 Germany
| | - Björn Braunschweig
- Institute of Physical Chemistry, Center for Soft Nanoscience (SoN), University of Münster Corrensstraße 28-30 Münster 48149 Germany
| |
Collapse
|
7
|
Chenab KK, Malektaj H, Nadinlooie AAR, Mohammadi S, Zamani-Meymian MR. Intertumoral and intratumoral barriers as approaches for drug delivery and theranostics to solid tumors using stimuli-responsive materials. Mikrochim Acta 2024; 191:541. [PMID: 39150483 DOI: 10.1007/s00604-024-06583-y] [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: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
The solid tumors provide a series of biological barriers in cellular microenvironment for designing drug delivery methods based on advanced stimuli-responsive materials. These intertumoral and intratumoral barriers consist of perforated endotheliums, tumor cell crowding, vascularity, lymphatic drainage blocking effect, extracellular matrix (ECM) proteins, hypoxia, and acidosis. Triggering opportunities have been drawn for solid tumor therapies based on single and dual stimuli-responsive drug delivery systems (DDSs) that not only improved drug targeting in deeper sites of the tumor microenvironments, but also facilitated the antitumor drug release efficiency. Single and dual stimuli-responsive materials which are known for their lowest side effects can be categorized in 17 main groups which involve to internal and external stimuli anticancer drug carriers in proportion to microenvironments of targeted solid tumors. Development of such drug carriers can circumvent barriers in clinical trial studies based on their superior capabilities in penetrating into more inaccessible sites of the tumor tissues. In recent designs, key characteristics of these DDSs such as fast response to intracellular and extracellular factors, effective cytotoxicity with minimum side effect, efficient permeability, and rate and location of drug release have been discussed as core concerns of designing paradigms of these materials.
Collapse
Affiliation(s)
- Karim Khanmohammadi Chenab
- Department of Chemistry, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
- Department of Physics, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, 9220, Aalborg, Denmark
| | | | | | | |
Collapse
|
8
|
Manteghi M, Can O, Kocagoz T. Peptosome: A New Efficient Transfection Tool as an Alternative to Liposome. Int J Mol Sci 2024; 25:6918. [PMID: 39000028 PMCID: PMC11241524 DOI: 10.3390/ijms25136918] [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: 04/30/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Gene therapy is one of the most promising techniques for treating genetic diseases and cancer. The current most important problem in gene therapy is gene delivery. Viral and non-viral vectors like liposomes, used for gene delivery, have many limitations. We have developed new hybrid peptides by combining cell-penetrating peptides (CPPs) with the DNA-binding domain of the human histone H4 protein. These small peptides bind to DNA molecules through their histone domain, leaving the CPP part free and available for binding and penetration into cells, forming complexes that we named "peptosomes". We evaluated the transfection efficiency of several hybrid peptides by delivering a plasmid carrying the green fluorescent protein gene and following its expression by fluorescent microscopy. Among several hybrid peptides, TM3 achieved a gene delivery efficiency of 76%, compared to 52% for Lipofectamine 2000. TM3 peptosomes may become important gene delivery tools with several advantages over current gene delivery agents.
Collapse
Affiliation(s)
- Maliheh Manteghi
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Ozge Can
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
| | - Tanil Kocagoz
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey;
- Department of Medical Microbiology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| |
Collapse
|
9
|
Mohammadi R, Ghani S, Arezumand R, Farhadi S, Khazaee-Poul Y, Kazemi B, Yarian F, Noruzi S, Alibakhshi A, Jalili M, Aghamiri S. Physicochemical Stimulus-Responsive Systems Targeted with Antibody Derivatives. Curr Mol Med 2024; 24:1250-1268. [PMID: 37594115 DOI: 10.2174/1566524023666230818093016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 08/19/2023]
Abstract
The application of monoclonal antibodies and antibody fragments with the advent of recombinant antibody technology has made notable progress in clinical trials to provide a regulated drug release and extra targeting to the special conditions in the function site. Modification of antibodies has facilitated using mAbs and antibody fragments in numerous models of therapeutic and detection utilizations, such as stimuliresponsive systems. Antibodies and antibody derivatives conjugated with diverse stimuliresponsive materials have been constructed for drug delivery in response to a wide range of endogenous (electric, magnetic, light, radiation, ultrasound) and exogenous (temperature, pH, redox potential, enzymes) stimuli. In this report, we highlighted the recent progress on antibody-conjugated stimuli-responsive and dual/multi-responsive systems that affect modern medicine by improving a multitude of diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Rezvan Mohammadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepideh Ghani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technology in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roghaye Arezumand
- Department of Advanced Technology, School of Medicine, North Khorasan University of Medical Sciences, North Khorasan, Iran
| | - Shohreh Farhadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yalda Khazaee-Poul
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Kazemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Yarian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Somaye Noruzi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Alibakhshi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahsa Jalili
- Preventive and Clinical Nutrition Group, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Shahin Aghamiri
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Uppalapati SS, Guha L, Kumar H, Mandoli A. Nanotechnological Advancements for the Theranostic Intervention in Anaplastic Thyroid Cancer: Current Perspectives and Future Direction. Curr Cancer Drug Targets 2024; 24:245-270. [PMID: 37424349 DOI: 10.2174/1568009623666230707155145] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/30/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023]
Abstract
Anaplastic thyroid cancer is the rarest, most aggressive, and undifferentiated class of thyroid cancer, accounting for nearly forty percent of all thyroid cancer-related deaths. It is caused by alterations in many cellular pathways like MAPK, PI3K/AKT/mTOR, ALK, Wnt activation, and TP53 inactivation. Although many treatment strategies, such as radiation therapy and chemotherapy, have been proposed to treat anaplastic thyroid carcinoma, they are usually accompanied by concerns such as resistance, which may lead to the lethality of the patient. The emerging nanotechnology-based approaches cater the purposes such as targeted drug delivery and modulation in drug release patterns based on internal or external stimuli, leading to an increase in drug concentration at the site of the action that gives the required therapeutic action as well as modulation in diagnostic intervention with the help of dye property materials. Nanotechnological platforms like liposomes, micelles, dendrimers, exosomes, and various nanoparticles are available and are of high research interest for therapeutic intervention in anaplastic thyroid cancer. The pro gression of the disease can also be traced by using magnetic probes or radio-labeled probes and quantum dots that serve as a diagnostic intervention in anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Sai Swetha Uppalapati
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, India
| | - Lahanya Guha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, India
| | - Amit Mandoli
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, India
| |
Collapse
|
11
|
Yan H, Xu P, Ma H, Li Y, Zhang R, Cong H, Yu B, Shen Y. Enzyme-triggered transcytosis of drug carrier system for deep penetration into hepatoma tumors. Biomaterials 2023; 301:122213. [PMID: 37385137 DOI: 10.1016/j.biomaterials.2023.122213] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 06/08/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
In recent years, nano-drug delivery systems have made considerable progress in the direction of tumor treatment, but the low permeability of drugs has restricted the development of nano drugs. To solve this problem, we constructed a nano-drug delivery system with the dual effects of γ-glutamyltransferase (GGT) reaction and high nuclear targeting in tumor microenvironment to promote the deep penetration of drugs. Over-expression of GGT in tumor cells can specifically recognize γ-glutamyl substrate and release amino group from the hydrolysis reaction, which makes the whole system change from negative or neutral to positive charge system. The conjugated complex with positive charge rapidly endocytosis through electrostatic interaction, enhancing its permeability in tumor parenchyma. At the same time, the cell penetrating TAT contains a large amount of lysine, which can be identified by the nuclear pore complexes (NPCs) on the surface of the nuclear membrane, showing excellent nuclear localization function. The active DOX is released in the nucleus, which inhibits the mitosis of cancer cells and enhances the active transport ability of drugs in tumor cells. Therefore, this drug delivery system actively transports adriamycin into the tumor to achieve deep penetration of drugs through enzyme response and nuclear targeting, showing high anti-tumor activity and can be effectively applied to the treatment of liver cancer.
Collapse
Affiliation(s)
- Han Yan
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Pengchao Xu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - He Ma
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Yanan Li
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Runfeng Zhang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, And Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
12
|
Tan KF, In LLA, Vijayaraj Kumar P. Surface Functionalization of Gold Nanoparticles for Targeting the Tumor Microenvironment to Improve Antitumor Efficiency. ACS APPLIED BIO MATERIALS 2023; 6:2944-2981. [PMID: 37435615 DOI: 10.1021/acsabm.3c00202] [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] [Indexed: 07/13/2023]
Abstract
Gold nanoparticles (AuNPs) have undergone significant research for their use in the treatment of cancer. Numerous researchers have established their potent antitumor properties, which have greatly impacted the treatment of cancer. AuNPs have been used in four primary anticancer treatment modalities, namely radiation, photothermal therapy, photodynamic therapy, and chemotherapy. However, the ability of AuNPs to destroy cancer is lacking and can even harm healthy cells without the right direction to transport them to the tumor microenvironment. Consequently, a suitable targeting technique is needed. Based on the distinct features of the human tumor microenvironment, this review discusses four different targeting strategies that target the four key features of the tumor microenvironment, including abnormal vasculature, overexpression of specific receptors, an acidic microenvironment, and a hypoxic microenvironment, to direct surface-functionalized AuNPs to the tumor microenvironment and increase antitumor efficacies. In addition, some current completed or ongoing clinical trials of AuNPs will also be discussed below to further reinforce the concept of using AuNPs in anticancer therapy.
Collapse
Affiliation(s)
- Kin Fai Tan
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| | - Lionel Lian Aun In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Palanirajan Vijayaraj Kumar
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, UCSI University, No. 1, Jalan Menara Gading, Taman Connaught, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
13
|
Zhang Z, Ding C, Sun T, Wang L, Chen C. Tumor Therapy Strategies Based on Microenvironment-Specific Responsive Nanomaterials. Adv Healthc Mater 2023; 12:e2300153. [PMID: 36933000 DOI: 10.1002/adhm.202300153] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/10/2023] [Indexed: 03/19/2023]
Abstract
The tumor microenvironment (TME) is a complex and variable region characterized by hypoxia, low pH, high redox status, overexpression of enzymes, and high-adenosine triphosphate concentrations. In recent years, with the continuous in-depth study of nanomaterials, more and more TME-specific response nanomaterials are used for tumor treatment. However, the complexity of the TME causes different types of responses with various strategies and mechanisms of action. Aiming to systematically demonstrate the recent advances in research on TME-responsive nanomaterials, this work summarizes the characteristics of TME and outlines the strategies of different TME responses. Representative reaction types are illustrated and their merits and demerits are analyzed. Finally, forward-looking views on TME-response strategies for nanomaterials are presented. It is envisaged that such emerging strategies for the treatment of cancer are expected to exhibit dramatic trans-clinical capabilities, demonstrating the extensive potential for the diagnosis and therapy of cancer.
Collapse
Affiliation(s)
- Zhaocong Zhang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Chengwen Ding
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Tiedong Sun
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Lei Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Chunxia Chen
- Key Laboratory of Forest Plant Ecology, Ministry of Education, College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| |
Collapse
|
14
|
Nel J, Elkhoury K, Velot É, Bianchi A, Acherar S, Francius G, Tamayol A, Grandemange S, Arab-Tehrany E. Functionalized liposomes for targeted breast cancer drug delivery. Bioact Mater 2023; 24:401-437. [PMID: 36632508 PMCID: PMC9812688 DOI: 10.1016/j.bioactmat.2022.12.027] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/05/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023] Open
Abstract
Despite the exceptional progress in breast cancer pathogenesis, prognosis, diagnosis, and treatment strategies, it remains a prominent cause of female mortality worldwide. Additionally, although chemotherapies are effective, they are associated with critical limitations, most notably their lack of specificity resulting in systemic toxicity and the eventual development of multi-drug resistance (MDR) cancer cells. Liposomes have proven to be an invaluable drug delivery system but of the multitudes of liposomal systems developed every year only a few have been approved for clinical use, none of which employ active targeting. In this review, we summarize the most recent strategies in development for actively targeted liposomal drug delivery systems for surface, transmembrane and internal cell receptors, enzymes, direct cell targeting and dual-targeting of breast cancer and breast cancer-associated cells, e.g., cancer stem cells, cells associated with the tumor microenvironment, etc.
Collapse
Affiliation(s)
- Janske Nel
- Université de Lorraine, LIBio, F-54000, Nancy, France
| | | | - Émilie Velot
- Université de Lorraine, CNRS, IMoPA, F-54000, Nancy, France
| | - Arnaud Bianchi
- Université de Lorraine, CNRS, IMoPA, F-54000, Nancy, France
| | - Samir Acherar
- Université de Lorraine, CNRS, LCPM, F-54000, Nancy, France
| | | | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | | | | |
Collapse
|
15
|
Meagher RB, Lewis ZA, Ambati S, Lin X. DectiSomes: C-type lectin receptor-targeted liposomes as pan-antifungal drugs. Adv Drug Deliv Rev 2023; 196:114776. [PMID: 36934519 PMCID: PMC10133202 DOI: 10.1016/j.addr.2023.114776] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023]
Abstract
Combatting the ever-increasing threat from invasive fungal pathogens faces numerous fundamental challenges, including constant human exposure to large reservoirs of species in the environment, the increasing population of immunocompromised or immunosuppressed individuals, the unsatisfactory efficacy of current antifungal drugs and their associated toxicity, and the scientific and economic barriers limiting a new antifungal pipeline. DectiSomes represent a new drug delivery platform that enhances antifungal efficacy for diverse fungal pathogens and reduces host toxicity for current and future antifungals. DectiSomes employ pathogen receptor proteins - C-type lectins - to target drug-loaded liposomes to conserved fungal cognate ligands and away from host cells. DectiSomes represent one leap forward for urgently needed effective pan-antifungal therapy. Herein, we discuss the problems of battling fungal diseases and the state of DectiSome development.
Collapse
Affiliation(s)
- Richard B Meagher
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Zachary A Lewis
- Department of Genetics, University of Georgia, Athens, GA 30602, USA; Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Suresh Ambati
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
16
|
Gyanani V, Goswami R. Key Design Features of Lipid Nanoparticles and Electrostatic Charge-Based Lipid Nanoparticle Targeting. Pharmaceutics 2023; 15:1184. [PMID: 37111668 PMCID: PMC10144967 DOI: 10.3390/pharmaceutics15041184] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/02/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Lipid nanoparticles (LNP) have gained much attention after the approval of mRNA COVID-19 vaccines. The considerable number of currently ongoing clinical studies are testament to this fact. These efforts towards the development of LNPs warrant an insight into the fundamental developmental aspects of such systems. In this review, we discuss the key design aspects that confer efficacy to a LNP delivery system, i.e., potency, biodegradability, and immunogenicity. We also cover the underlying considerations regarding the route of administration and targeting of LNPs to hepatic and non-hepatic targets. Furthermore, since LNP efficacy is also a function of drug/nucleic acid release within endosomes, we take a holistic view of charged-based targeting approaches of LNPs not only in the context of endosomal escape but also in relation to other comparable target cell internalization strategies. Electrostatic charge-based interactions have been used in the past as a potential strategy to enhance the drug release from pH-sensitive liposomes. In this review, we cover such strategies around endosomal escape and cell internalization in low pH tumor micro-environments.
Collapse
Affiliation(s)
- Vijay Gyanani
- T.J.L. School of Pharmacy, University of the Pacific, Stockton, CA 95211, USA
| | | |
Collapse
|
17
|
Shadmani N, Makvandi P, Parsa M, Azadi A, Nedaei K, Mozafari N, Poursina N, Mattoli V, Tay FR, Maleki A, Hamidi M. Enhancing Methotrexate Delivery in the Brain by Mesoporous Silica Nanoparticles Functionalized with Cell-Penetrating Peptide using in Vivo and ex Vivo Monitoring. Mol Pharm 2023; 20:1531-1548. [PMID: 36763486 DOI: 10.1021/acs.molpharmaceut.2c00755] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The blood-brain barrier (BBB) acts as a physical/biochemical barrier that protects brain parenchyma from potential hazards exerted by different xenobiotics found in the systemic circulation. This barrier is created by "a lipophilic gate" as well as a series of highly organized influx/efflux mechanisms. The BBB bottleneck adversely affects the efficacy of chemotherapeutic agents in treating different CNS malignancies such as glioblastoma, an aggressive type of cancer affecting the brain. In the present study, mesoporous silica nanoparticles (MSNs) were conjugated with the transactivator of transcription (TAT) peptide, a cell-penetrating peptide, to produce MSN-NH-TAT with the aim of improving methotrexate (MTX) penetration into the brain. The TAT-modified nanosystem was characterized by Fourier transform infrared spectrometry (FTIR), field emission scanning electron microscopy (FE-SEM), transmission electron microscopy (TEM), atomic force microscopy (AFM), dynamic light scattering (DLS), and N2 adsorption-desorption analysis. In vitro hemolysis and cell viability studies confirmed the biocompatibility of the MSN-based nanocarriers. In addition, in vivo studies showed that the MTX-loaded MSN-NH-TAT improved brain-to-plasma concentration ratio, brain uptake clearance, and the drug's blood terminal half-life, compared with the use of free MTX. Taken together, the results of the present study indicate that MSN functionalization with TAT is crucial for delivery of MTX into the brain. The present nanosystem represents a promising alternative drug carrier to deliver MTX into the brain via overcoming the BBB.
Collapse
Affiliation(s)
- Nasim Shadmani
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran.,Trita Nanomedicine Research & Technology Development Center (TNRTC), Zanjan Health Technology Park, 45156-13191Zanjan, Iran
| | - Pooyan Makvandi
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, EdinburghEH9 3JL, U.K
| | - Maliheh Parsa
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran
| | - Amir Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685Shiraz, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, 71468 64685Shiraz, Iran
| | - Keivan Nedaei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran
| | - Negin Mozafari
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685Shiraz, Iran
| | - Narges Poursina
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran
| | - Virgilio Mattoli
- Centre for Materials Interfaces, Istituto Italiano di Tecnologia, Viale Rinaldo Piaggio 34, 56025Pontedera, Pisa, Italy
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, Georgia30912, United States
| | - Aziz Maleki
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran
| | - Mehrdad Hamidi
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran.,Trita Nanomedicine Research & Technology Development Center (TNRTC), Zanjan Health Technology Park, 45156-13191Zanjan, Iran.,Department of Pharmaceutics, School of Pharmacy, Zanjan University of Medical Sciences, 45139-56184Zanjan, Iran
| |
Collapse
|
18
|
Spleis H, Sandmeier M, Claus V, Bernkop-Schnürch A. Surface design of nanocarriers: Key to more efficient oral drug delivery systems. Adv Colloid Interface Sci 2023; 313:102848. [PMID: 36780780 DOI: 10.1016/j.cis.2023.102848] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
As nanocarriers (NCs) can improve the solubility of drugs, prevent their degradation by gastrointestinal (GI) enzymes and promote their transport across the mucus gel layer and absorption membrane, the oral bioavailability of these drugs can be substantially enhanced. All these properties of NCs including self-emulsifying drug delivery systems (SEDDS), solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), liposomes, polymeric nanoparticles, inorganic nanoparticles and polymeric micelles depend mainly on their surface chemistry. In particular, interaction with food, digestive enzymes, bile salts and electrolytes, diffusion behaviour across the mucus gel layer and fate on the absorption membrane are determined by their surface. Bioinert surfaces limiting interactions with gastrointestinal fluid and content as well as with mucus, adhesive surfaces providing an intimate contact with the GI mucosa and absorption enhancing surfaces can be designed. Furthermore, charge converting surfaces shifting their zeta potential from negative to positive directly at the absorption membrane and surfaces providing a targeted drug release are advantageous. In addition to these passive surfaces, even active surfaces cleaving mucus glycoproteins on their way through the mucus gel layer can be created. Within this review, we provide an overview on these different surfaces and discuss their impact on the performance of NCs in the GI tract.
Collapse
Affiliation(s)
- Helen Spleis
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Matthias Sandmeier
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Victor Claus
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria; Thiomatrix Forschungs und Beratungs GmbH, Trientlgasse 65, Innsbruck 6020, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck 6020, Austria.
| |
Collapse
|
19
|
He N, Wang A, Tian C, Song Y, Guo X, Ming H, Ding M, Luo F, Tan H, Li J. Tuning the Endocytosis of Hybrid Micelles through Spatial Regulation of Cationic Groups. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 36779657 DOI: 10.1021/acsami.2c20620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The ability of nanocarriers to enter tumor cells can be enhanced by positive surface charge. Nonetheless, the relationship between the spatial distributions of cationic groups and the endocytosis and tumor penetration of nanocarriers remains largely elusive. Here, using quaternary ammonium salt (QAS) as a model cationic group, a series of hybrid micelles (HMs) bearing QAS with different spatial distributions were prepared from star-shaped polymers with well-defined molecular architectures. The structural characteristics of HM, such as spatial location of QAS and local poly(ethylene glycol) (PEG) density near QAS, were investigated by both experimental techniques and dissipative particle dynamics (DPD) simulation. We show that the drug carriers with QAS extending to the micellar outer space allows QAS to facilitate cell surface binding with minimized hindrance, resulting in greatly enhanced endocytosis compared with nanocarriers with QAS attached onto the micellar surface or shielded by a PEG corona. This study offers cues for future development of tumor-penetrating drug delivery systems.
Collapse
Affiliation(s)
- Nan He
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Ao Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Chenxu Tian
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Yuanqing Song
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Xiaolei Guo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hao Ming
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Mingming Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Feng Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Jiehua Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
20
|
Marciano Y, del Solar V, Nayeem N, Dave D, Son J, Contel M, Ulijn RV. Encapsulation of Gold-Based Anticancer Agents in Protease-Degradable Peptide Nanofilaments Enhances Their Potency. J Am Chem Soc 2023; 145:234-246. [PMID: 36542079 PMCID: PMC10720394 DOI: 10.1021/jacs.2c09820] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We investigated the use of amphiphilic, protease-cleavable peptides as encapsulation moieties for hydrophobic metallodrugs, in order to enhance their bioavailability and consequent activity. Two hydrophobic, gold-containing anticancer agents varying in aromatic ligand distribution (Au(I)-N-heterocyclic carbene compounds 1 and 2) were investigated. These were encapsulated into amphiphilic decapeptides that form soluble filamentous structures with hydrophobic cores, varying supramolecular packing arrangements and surface charge. Peptide sequence strongly dictates the supramolecular packing within the aromatic core, which in turn dictates drug loading. Anionic peptide filaments can effectively load 1, and to a lesser extent 2, while their cationic counterparts could not, collectively demonstrating that loading efficiency is dictated by both aromatic and electrostatic (mis)matching between drug and peptide. Peptide nanofilaments were nontoxic to cancerous and noncancerous cells. By contrast, those loaded with 1 and 2 displayed enhanced cytotoxicity in comparison to 1 and 2 alone, when exposed to Caki-1 and MDA-MB-231 cancerous cell lines, while no cytotoxicity was observed in noncancerous lung fibroblasts, IMR-90. We propose that the enhanced in vitro activity results from the enhanced proteolytic activity in the vicinity of the cancer cells, thereby breaking the filaments into drug-bound peptide fragments that are taken up by these cells, resulting in enhanced cytotoxicity toward cancer cells.
Collapse
Affiliation(s)
- Yaron Marciano
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Ph.D. Program in Chemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
| | - Virginia del Solar
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
| | - Nazia Nayeem
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Ph.D. Program inBiology, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
| | - Dhwanit Dave
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA
- Ph.D. Program in Chemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Department of Chemistry, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| | - Jiye Son
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA
| | - María Contel
- Department of Chemistry, Brooklyn College, CUNY, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Ph.D. Program in Chemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Ph.D. Program in Biochemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Ph.D. Program inBiology, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
| | - Rein V. Ulijn
- Nanoscience Initiative, Advanced Science Research Center at The Graduate Center of the City University of New York (CUNY), 85 Saint Nicholas Terrace, New York, NY 10031, USA
- Ph.D. Program in Chemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Ph.D. Program in Biochemistry, The Graduate Center of CUNY, 365 Fifth Avenue, New York, NY 10016, USA
- Department of Chemistry, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA
| |
Collapse
|
21
|
Rana A, Adhikary M, Singh PK, Das BC, Bhatnagar S. "Smart" drug delivery: A window to future of translational medicine. Front Chem 2023; 10:1095598. [PMID: 36688039 PMCID: PMC9846181 DOI: 10.3389/fchem.2022.1095598] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/28/2022] [Indexed: 01/05/2023] Open
Abstract
Chemotherapy is the mainstay of cancer treatment today. Chemotherapeutic drugs are non-selective and can harm both cancer and healthy cells, causing a variety of adverse effects such as lack of specificity, cytotoxicity, short half-life, poor solubility, multidrug resistance, and acquiring cancer stem-like characteristics. There is a paradigm shift in drug delivery systems (DDS) with the advent of smarter ways of targeted cancer treatment. Smart Drug Delivery Systems (SDDSs) are stimuli responsive and can be modified in chemical structure in response to light, pH, redox, magnetic fields, and enzyme degradation can be future of translational medicine. Therefore, SDDSs have the potential to be used as a viable cancer treatment alternative to traditional chemotherapy. This review focuses mostly on stimuli responsive drug delivery, inorganic nanocarriers (Carbon nanotubes, gold nanoparticles, Meso-porous silica nanoparticles, quantum dots etc.), organic nanocarriers (Dendrimers, liposomes, micelles), antibody-drug conjugates (ADC) and small molecule drug conjugates (SMDC) based SDDSs for targeted cancer therapy and strategies of targeted drug delivery systems in cancer cells.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Meheli Adhikary
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Praveen Kumar Singh
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Bhudev C. Das
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India,Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India,*Correspondence: Seema Bhatnagar,
| |
Collapse
|
22
|
Hashemi M, Ghadyani F, Hasani S, Olyaee Y, Raei B, Khodadadi M, Ziyarani MF, Basti FA, Tavakolpournegari A, Matinahmadi A, Salimimoghadam S, Aref AR, Taheriazam A, Entezari M, Ertas YN. Nanoliposomes for doxorubicin delivery: Reversing drug resistance, stimuli-responsive carriers and clinical translation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
23
|
Ashrafizadeh M, Delfi M, Zarrabi A, Bigham A, Sharifi E, Rabiee N, Paiva-Santos AC, Kumar AP, Tan SC, Hushmandi K, Ren J, Zare EN, Makvandi P. Stimuli-responsive liposomal nanoformulations in cancer therapy: Pre-clinical & clinical approaches. J Control Release 2022; 351:50-80. [PMID: 35934254 DOI: 10.1016/j.jconrel.2022.08.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
The site-specific delivery of antitumor agents is of importance for providing effective cancer suppression. Poor bioavailability of anticancer compounds and the presence of biological barriers prevent their accumulation in tumor sites. These obstacles can be overcome using liposomal nanostructures. The challenges in cancer chemotherapy and stimuli-responsive nanocarriers are first described in the current review. Then, stimuli-responsive liposomes including pH-, redox-, enzyme-, light-, thermo- and magneto-sensitive nanoparticles are discussed and their potential for delivery of anticancer drugs is emphasized. The pH- or redox-sensitive liposomes are based on internal stimulus and release drug in response to a mildly acidic pH and GSH, respectively. The pH-sensitive liposomes can mediate endosomal escape via proton sponge. The multifunctional liposomes responsive to both redox and pH have more capacity in drug release at tumor site compared to pH- or redox-sensitive alone. The magnetic field and NIR irradiation can be exploited for external stimulation of liposomes. The light-responsive liposomes release drugs when they are exposed to irradiation; thermosensitive-liposomes release drugs at a temperature of >40 °C when there is hyperthermia; magneto-responsive liposomes release drugs in presence of magnetic field. These smart nanoliposomes also mediate co-delivery of drugs and genes in synergistic cancer therapy. Due to lack of long-term toxicity of liposomes, they can be utilized in near future for treatment of cancer patients.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey.
| | - Masoud Delfi
- Department of Chemical Sciences, University of Naples "Federico II", Complesso Universitario Monte S. Angelo, Via Cintia, Naples 80126, Italy
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials-National Research Council (IPCB-CNR), Viale J.F. Kennedy 54-Mostra d'Oltremare pad. 20, 80125 Naples, Italy
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Navid Rabiee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, South Korea; School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology and zoonosis, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | | | - Pooyan Makvandi
- School of Chemistry, Damghan University, Damghan 36716-41167, Iran; Istituto Italiano di Tecnologia, Center for Materials Interfaces, viale Rinaldo Piaggio 34, 56025, Pontedera, Pisa, Italy.
| |
Collapse
|
24
|
Zhang Z, Gao A, Sun C. Tumor pH-Responsive Nanocarriers With Light-Activatable Drug Release for Chemo-Photodynamic Therapy of Breast Cancer. Front Chem 2022; 10:905645. [PMID: 35815218 PMCID: PMC9257215 DOI: 10.3389/fchem.2022.905645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
Developing bioresponsive nanocarriers with particular tumor cell targeting and on-demand payload release has remained a great challenge for combined chemo-photodynamic therapy (chemo-PDT). In this study, an intelligent nanocarrier (DATAT-NPCe6) responded to hierarchical endogenous tumor pH, and an exogenous red light was developed through a simple mixed micelle approach. The outside TAT ligand was masked to prevent an unexpected interaction in blood circulation. Following the accumulation of DATAT-NPCe6 in tumor tissues, tumor acidity at pH ∼6.5 recovered its targeting ability via triggering DA moiety degradation. Furthermore, the cascaded chemo-PDT was accomplished through light-stimulated nanocarrier disassembly and doxorubicin (DOX) release. Taking advantage of stability and controllability, this work provides a facile approach to designing bioresponsive nanocarriers and represents a proof-of-concept combinatorial chemo-PDT treatment.
Collapse
|
25
|
Tian Y, Tirrell MV, LaBelle JL. Harnessing the Therapeutic Potential of Biomacromolecules through Intracellular Delivery of Nucleic Acids, Peptides, and Proteins. Adv Healthc Mater 2022; 11:e2102600. [PMID: 35285167 PMCID: PMC9232950 DOI: 10.1002/adhm.202102600] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/09/2022] [Indexed: 12/19/2022]
Abstract
Biomacromolecules have long been at the leading edge of academic and pharmaceutical drug development and clinical translation. With the clinical advances of new therapeutics, such as monoclonal antibodies and nucleic acids, the array of medical applications of biomacromolecules has broadened considerably. A major on-going effort is to expand therapeutic targets within intracellular locations. Owing to their large sizes, abundant charges, and hydrogen-bond donors and acceptors, advanced delivery technologies are required to deliver biomacromolecules effectively inside cells. In this review, strategies used for the intracellular delivery of three major forms of biomacromolecules: nucleic acids, proteins, and peptides, are highlighted. An emphasis is placed on synthetic delivery approaches and the major hurdles needed to be overcome for their ultimate clinical translation.
Collapse
Affiliation(s)
- Yu Tian
- Pritzker School of Molecular EngineeringThe University of Chicago5640 S Ellis AveChicagoIL60637USA
| | - Matthew V. Tirrell
- Pritzker School of Molecular EngineeringThe University of Chicago5640 S Ellis AveChicagoIL60637USA
| | - James L. LaBelle
- Department of Pediatrics, Section of Hematology/OncologyThe University of Chicago900 E 57th StChicagoIL60637USA
| |
Collapse
|
26
|
Nsairat H, Khater D, Sayed U, Odeh F, Al Bawab A, Alshaer W. Liposomes: structure, composition, types, and clinical applications. Heliyon 2022; 8:e09394. [PMID: 35600452 PMCID: PMC9118483 DOI: 10.1016/j.heliyon.2022.e09394] [Citation(s) in RCA: 427] [Impact Index Per Article: 142.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/19/2022] [Accepted: 05/06/2022] [Indexed: 12/18/2022] Open
Abstract
Liposomes are now considered the most commonly used nanocarriers for various potentially active hydrophobic and hydrophilic molecules due to their high biocompatibility, biodegradability, and low immunogenicity. Liposomes also proved to enhance drug solubility and controlled distribution, as well as their capacity for surface modifications for targeted, prolonged, and sustained release. Based on the composition, liposomes can be considered to have evolved from conventional, long-circulating, targeted, and immune-liposomes to stimuli-responsive and actively targeted liposomes. Many liposomal-based drug delivery systems are currently clinically approved to treat several diseases, such as cancer, fungal and viral infections; more liposomes have reached advanced phases in clinical trials. This review describes liposomes structure, composition, preparation methods, and clinical applications.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Dima Khater
- Department of Chemistry, Faculty of Arts and Science, Applied Science Private University, Amman, Jordan
| | - Usama Sayed
- Department of Biology, The University of Jordan, Amman, 11942, Jordan
| | - Fadwa Odeh
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan
| | - Abeer Al Bawab
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.,Hamdi Mango Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
27
|
Zhou M, Zou X, Cheng K, Zhong S, Su Y, Wu T, Tao Y, Cong L, Yan B, Jiang Y. The role of cell-penetrating peptides in potential anti-cancer therapy. Clin Transl Med 2022; 12:e822. [PMID: 35593206 PMCID: PMC9121317 DOI: 10.1002/ctm2.822] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 12/19/2022] Open
Abstract
Due to the complex physiological structure, microenvironment and multiple physiological barriers, traditional anti-cancer drugs are severely restricted from reaching the tumour site. Cell-penetrating peptides (CPPs) are typically made up of 5-30 amino acids, and can be utilised as molecular transporters to facilitate the passage of therapeutic drugs across physiological barriers. Up to now, CPPs have widely been used in many anti-cancer treatment strategies, serving as an excellent potential choice for oncology treatment. However, their drawbacks, such as the lack of cell specificity, short duration of action, poor stability in vivo, compatibility problems (i.e. immunogenicity), poor therapeutic efficacy and formation of unwanted metabolites, have limited their further application in cancer treatment. The cellular uptake mechanisms of CPPs involve mainly endocytosis and direct penetration, but still remain highly controversial in academia. The CPPs-based drug delivery strategy could be improved by clever design or chemical modifications to develop the next-generation CPPs with enhanced cell penetration capability, stability and selectivity. In addition, some recent advances in targeted cell penetration that involve CPPs provide some new ideas to optimise CPPs.
Collapse
Affiliation(s)
- Meiling Zhou
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Xi Zou
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Kexin Cheng
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Suye Zhong
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Yangzhou Su
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Tao Wu
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Li Cong
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Bin Yan
- Department of Pathology, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Yiqun Jiang
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha, Hunan, China.,School of Medicine, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
28
|
Cao Y, Zhang S, Ma M, Zhang Y. Fluorinated PEG-PEI Coated Magnetic Nanoparticles for siRNA Delivery and CXCR4 Knockdown. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1692. [PMID: 35630915 PMCID: PMC9146302 DOI: 10.3390/nano12101692] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 02/08/2023]
Abstract
CXC chemokine receptor 4 (CXCR4) is a promising therapeutic target. Previous studies have shown that intracellular delivery of siRNA to knockdown CXCR4 expression in cancer cells is an effective therapeutic strategy. To prepare efficient magnetic nucleic acid carriers, it is now necessary to improve the endocytosis efficiency of PEGylated magnetic nanoparticles. In our work, Heptafluorobutyryl-polyethylene glycol-polyethyleneimine (FPP) was first prepared and then used to coat magnetic nanoparticles (MNPs) to obtain magnetic nanocarriers FPP@MNPs. The materials were characterized by 19 F-Nuclear Magnetic Resonance (NMR), transmission electron microscope (TEM), energy dispersive spectroscopy (EDS), and dynamic light scattering (DLS). The biosecurity of FPP@MNPs was confirmed by cell viability and apoptosis experiments. Cellular uptake of FPP@MNPs and siRNA transfection enhanced by external magnetic fields were detected by fluorescence microscopy, confocal laser microscopy, and flow cytometry. The results show that the cellular uptake efficiency of FPP@MNPs was significantly improved, and transfection efficiency reached more than 90%. The knockdown of CXCR4 on the 4 T1 cell membrane was confirmed by real-time polymerase chain reaction (RT-PCR) and flow cytometry. In conclusion, the fluorinated cationic polymer-coated magnetic nanoparticles FPP@MNPs can be loaded with siRNA to reduce CXCR4 expression as well as be expected to be efficient universal siRNA carriers.
Collapse
Affiliation(s)
- Yixiang Cao
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China;
| | - Shiyin Zhang
- Nanjing Nanoeast Biotech Co., Ltd., Nanjing 211000, China;
| | - Ming Ma
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China;
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing 210096, China;
| |
Collapse
|
29
|
Wang X, Wu C, Liu S, Peng D. Combinatorial therapeutic strategies for enhanced delivery of therapeutics to brain cancer cells through nanocarriers: current trends and future perspectives. Drug Deliv 2022; 29:1370-1383. [PMID: 35532094 PMCID: PMC9090367 DOI: 10.1080/10717544.2022.2069881] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Brain cancer is the most aggressive one among various cancers. It has a drastic impact on people's lives because of the failure in treatment efficacy of the currently employed strategies. Various strategies used to relieve pain in brain cancer patients and to prolong survival time include radiotherapy, chemotherapy, and surgery. Nevertheless, several inevitable limitations are accompanied by such treatments due to unsatisfactory curative effects. Generally, the treatment of cancers is very challenging due to many reasons including drugs’ intrinsic factors and physiological barriers. Blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB) are the two additional hurdles in the way of therapeutic agents to brain tumors delivery. Combinatorial and targeted therapies specifically in cancer show a very promising role where nanocarriers’ based formulations are designed primarily to achieve tumor-specific drug release. A dual-targeting strategy is a versatile way of chemotherapeutics delivery to brain tumors that gets the aid of combined ligands and mediators that cross the BBB and reaches the target site efficiently. In contrast to single targeting where one receptor or mediator is targeted, the dual-targeting strategy is expected to produce a multiple-fold increase in therapeutic efficacy for cancer therapy, especially in brain tumors. In a nutshell, a dual-targeting strategy for brain tumors enhances the delivery efficiency of chemotherapeutic agents via penetration across the blood-brain barrier and enhances the targeting of tumor cells. This review article highlights the ongoing status of the brain tumor therapy enhanced by nanoparticle based delivery with the aid of dual-targeting strategies. The future perspectives in this regard have also been highlighted.
Collapse
Affiliation(s)
- Xiande Wang
- Department of Neurosurgery, Hangzhou Medical College Affiliated Lin'an People's Hospital, The First People's Hospital of Hangzhou Lin'an District, Hangzhou, China
| | - Cheng Wu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Shiming Liu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Deqing Peng
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
30
|
Chu S, Shi X, Tian Y, Gao F. pH-Responsive Polymer Nanomaterials for Tumor Therapy. Front Oncol 2022; 12:855019. [PMID: 35392227 PMCID: PMC8980858 DOI: 10.3389/fonc.2022.855019] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
The complexity of the tumor microenvironment presents significant challenges to cancer therapy, while providing opportunities for targeted drug delivery. Using characteristic signals of the tumor microenvironment, various stimuli-responsive drug delivery systems can be constructed for targeted drug delivery to tumor sites. Among these, the pH is frequently utilized, owing to the pH of the tumor microenvironment being lower than that of blood and healthy tissues. pH-responsive polymer carriers can improve the efficiency of drug delivery in vivo, allow targeted drug delivery, and reduce adverse drug reactions, enabling multifunctional and personalized treatment. pH-responsive polymers have gained increasing interest due to their advantageous properties and potential for applicability in tumor therapy. In this review, recent advances in, and common applications of, pH-responsive polymer nanomaterials for drug delivery in cancer therapy are summarized, with a focus on the different types of pH-responsive polymers. Moreover, the challenges and future applications in this field are prospected.
Collapse
Affiliation(s)
- Shunli Chu
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiaolu Shi
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Ye Tian
- Department of Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Fengxiang Gao
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
31
|
Jing X, Hu H, Sun Y, Yu B, Cong H, Shen Y. The Intracellular and Extracellular Microenvironment of Tumor Site: The Trigger of Stimuli-Responsive Drug Delivery Systems. SMALL METHODS 2022; 6:e2101437. [PMID: 35048560 DOI: 10.1002/smtd.202101437] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/29/2021] [Indexed: 06/14/2023]
Abstract
The tumor microenvironment (TME), including intracellular and extracellular microenvironment, contains many biochemical indicators (such as acidity/alkalinity, oxygen content, and enzymatic activity) that are different from the normal physiological environment. These abnormal biochemical indicators can accelerate the heterogeneity of tumors, but on the other hand, they also provide opportunities for the design of intelligent drug delivery systems (DDSs). The TME-responsive DDSs have shown great potential in reducing the side effects of chemotherapy and improving the curative effect of tumors. In this review, the abnormal biochemical indicators of TME are introduced in detail from both the extracellular and intracellular aspects. In view of the various physiological barriers encountered during drug delivery, the strategy of constructing TME-responsive DDSs is discussed. By summarizing the typical research progress, the authors prospect the development of TME-responsive DDS in the future.
Collapse
Affiliation(s)
- Xiaodong Jing
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Hao Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Yanzhen Sun
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
32
|
Enzyme-sensitive nanoparticles, smart TAT and cetuximab conjugated immunoliposomes to overcome multidrug resistance in breast cancer cells. Toxicol Appl Pharmacol 2022; 441:115989. [DOI: 10.1016/j.taap.2022.115989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 11/15/2022]
|
33
|
Chen Q, Li Q, Liang Y, Zu M, Chen N, Canup BS, Luo L, Wang C, Zeng L, Xiao B. Natural exosome-like nanovesicles from edible tea flowers suppress metastatic breast cancer via ROS generation and microbiota modulation. Acta Pharm Sin B 2022; 12:907-923. [PMID: 35256954 PMCID: PMC8897038 DOI: 10.1016/j.apsb.2021.08.016] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/22/2021] [Accepted: 07/05/2021] [Indexed: 12/17/2022] Open
Abstract
Although several artificial nanotherapeutics have been approved for practical treatment of metastatic breast cancer, their inefficient therapeutic outcomes, serious adverse effects, and high cost of mass production remain crucial challenges. Herein, we developed an alternative strategy to specifically trigger apoptosis of breast tumors and inhibit their lung metastasis by using natural nanovehicles from tea flowers (TFENs). These nanovehicles had desirable particle sizes (131 nm), exosome-like morphology, and negative zeta potentials. Furthermore, TFENs were found to contain large amounts of polyphenols, flavonoids, functional proteins, and lipids. Cell experiments revealed that TFENs showed strong cytotoxicities against cancer cells due to the stimulation of reactive oxygen species (ROS) amplification. The increased intracellular ROS amounts could not only trigger mitochondrial damage, but also arrest cell cycle, resulting in the in vitro anti-proliferation, anti-migration, and anti-invasion activities against breast cancer cells. Further mice investigations demonstrated that TFENs after intravenous (i.v.) injection or oral administration could accumulate in breast tumors and lung metastatic sites, inhibit the growth and metastasis of breast cancer, and modulate gut microbiota. This study brings new insights to the green production of natural exosome-like nanoplatform for the inhibition of breast cancer and its lung metastasis via i.v. and oral routes.
Collapse
Key Words
- AF633, Alexa Fluor 633-labeled phalloidin
- ALP, alkaline phosphatase
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- BUN, urea nitrogen
- Breast cancer
- CDK, CYCLIN-dependent kinase
- CRE, creatinine
- DAF-FM DA, 4-amino-5-methylamino-2′,7′-difluorofluorescein diacetate
- DAPI, 4′,6-diamidino-2-phenylindole
- DCFH-DA, dichloro-dihydro-fluorescein diacetate
- DGDG, digalactosyl diacylglycerols
- DHE, dihydroethidium
- DLS, dynamic light scattering
- DiO, 3,3′-dioctadecyloxacarbocyanine perchlorate
- DiR, 1,1′-dioctadecyl-3,3,3′′,3′-tetramethylindotricarbocyanine iodide
- EC, epicatechin
- ECG, epicatechin gallate
- EGCG, epigallocatechin gallate
- Exosome-like nanoparticle
- FBS, fetal bovine serum
- GIT, gastrointestinal tract
- H&E, Hematoxylin & Eosin
- HPLC, high-performance liquid chromatography
- Intravenous injection
- LC‒MS, liquid chromatography‒mass spectrometry
- MFI, mean fluorescence intensity
- MGDG, monogalactosyl diacylglycerols
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- Metastasis
- Microbiota modulation
- NO, nitrogen monoxide
- NPs, nanoparticles
- OUT, operational taxonomic unit
- Oral administration
- PA, phosphatidic acids
- PBS, phosphate-buffered saline
- PC, phosphatidylcholines
- PDI, polydispersity index
- PE, phosphatidylethanolamines
- PG, phosphatidylglycerol
- PI, phosphatidylinositol
- PLT, platelets
- PMe, phosphatidylmethanol
- PS, phosphatidylserine
- RBC, red blood cell
- RNS, reactive nitrogen species
- ROS generation
- ROS, reactive oxygen species
- SA, superoxide anion
- SQDG, sulphoquinovosyl diylyceride
- TEM, transmission electron microscopy
- TFENs, exosome-like NPs from tea flowers
- TG, triglyceride
- TUNEL, TdT-mediated dUTP Nick-end labeling
- Tea flower
- WBC, white blood cell
Collapse
|
34
|
Chen S, Morrison G, Liu W, Kaur A, Chen R. A pH-responsive, endosomolytic liposome functionalized with membrane-anchoring, comb-like pseudopeptides for enhanced intracellular delivery and cancer treatment. Biomater Sci 2022; 10:6718-6730. [DOI: 10.1039/d2bm01087a] [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
Low intracellular delivery efficiency and multidrug resistance are among major barriers to effective cancer therapy.
Collapse
Affiliation(s)
- Siyuan Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing 212000, China
| | - Gabriella Morrison
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Wenyuan Liu
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Suqian Advanced Materials Industry Technology Innovation Center, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing 212000, China
| | - Apanpreet Kaur
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Rongjun Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| |
Collapse
|
35
|
Li L, Zhan Q, Yi K, Chen N, Li X, Yang S, Hou X, Zhao J, Yuan X, Kang C. Engineering Lipusu by lysophosphatidylcholine for improved tumor cellular uptake and anticancer efficacy. J Mater Chem B 2022; 10:1833-1842. [DOI: 10.1039/d1tb02823e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Liposomes have been developed as drug delivery carriers to enhance the antitumor efficiency of the therapeutic agents. Lipusu® (Lip), a paclitaxel (PTX) liposome, has been widely used in the treatment...
Collapse
|
36
|
Potential of cell-penetrating peptides (CPPs) in delivery of antiviral therapeutics and vaccines. Eur J Pharm Sci 2021; 169:106094. [PMID: 34896590 DOI: 10.1016/j.ejps.2021.106094] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
Viral infections are a great threat to human health. Currently, there are no effective vaccines and antiviral drugs against the majority of viral diseases, suggesting the need to develop novel and effective antiviral agents. Since the intracellular delivery of antiviral agents, particularly the impermeable molecules, such as peptides, proteins, and nucleic acids, are essential to exert their therapeutic effects, using a delivery system is highly required. Among various delivery systems, cell-penetrating peptides (CPPs), a group of short peptides with the unique ability of crossing cell membrane, offer great potential for the intracellular delivery of various biologically active cargoes. The results of numerous in vitro and in vivo studies with CPP conjugates demonstrate their promise as therapeutic agents in various medical fields including antiviral therapy. The CPP-mediated delivery of various antiviral agents including peptides, proteins, nucleic acids, and nanocarriers have been associated with therapeutic efficacy both in vitro and in vivo. This review describes various aspects of viruses including their biology, pathogenesis, and therapy and briefly discusses the concept of CPP and its potential in drug delivery. Particularly, it will highlight a variety of CPP applications in the management of viral infections.
Collapse
|
37
|
Dai J, Dong X, Wang Q, Lou X, Xia F, Wang S. PEG-Polymer Encapsulated Aggregation-Induced Emission Nanoparticles for Tumor Theranostics. Adv Healthc Mater 2021; 10:e2101036. [PMID: 34414687 DOI: 10.1002/adhm.202101036] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/28/2021] [Indexed: 12/15/2022]
Abstract
In the field of tumor imaging and therapy, the aggregation-caused quenching (ACQ) effect of fluorescent dyes at high concentration is a great challenge. In this regard, the aggregation-induced emission luminogens (AIEgens) show great potential, since AIEgens effectively overcome the ACQ effect and have better fluorescence quantum yield, photobleaching resistance, and photosensitivity. Polyethylene glycol (PEG)-polymer is the most commonly used carrier to prepare nanoparticles (NPs). The advantage of PEGylation is that it can greatly prolong the metabolic half-life and reduce immunogenicity and toxicity. Considering that the hydrophobicity of most AIEgens hinders their application in organisms, the use of PEG-polymer encapsulation is an effective strategy to overcome this obstacle. Importantly, bioactive functional groups can be modified on PEG-polymers to enhance the biological effect of NPs. The combination of powerful AIEgens and PEG-polymers provides a new strategy for tumor imaging and therapy, which is promising for clinical application.
Collapse
Affiliation(s)
- Jun Dai
- Department of Obstetrics and Gynecology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology 1095 Jiefang Avenue Wuhan 430032 China
| | - Xiaoqi Dong
- State Key Laboratory of Biogeology and Environmental Geology Faculty of Materials Science and Chemistry China University of Geosciences 388 Lumo Road Wuhan 430074 China
| | - Quan Wang
- State Key Laboratory of Biogeology and Environmental Geology Faculty of Materials Science and Chemistry China University of Geosciences 388 Lumo Road Wuhan 430074 China
| | - Xiaoding Lou
- State Key Laboratory of Biogeology and Environmental Geology Faculty of Materials Science and Chemistry China University of Geosciences 388 Lumo Road Wuhan 430074 China
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental Geology Faculty of Materials Science and Chemistry China University of Geosciences 388 Lumo Road Wuhan 430074 China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology 1095 Jiefang Avenue Wuhan 430032 China
| |
Collapse
|
38
|
Deng M, Rao JD, Guo R, Li M, He Q. Size-Adjustable Nano-Drug Delivery Systems for Enhanced Tumor Retention and Penetration. PHARMACEUTICAL FRONTS 2021. [DOI: 10.1055/s-0041-1736474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Over the past decades, nano-drug delivery systems have shown great potential in improving tumor treatment. And the controllability and design flexibility of nanoparticles endow them a broad development space. The particle size is one of the most important factors affecting the potency of nano-drug delivery systems. Large-size (100–200 nm) nanoparticles are more conducive to long circulation and tumor retention, but have poor tumor penetration; small-size (<50 nm) nanoparticles can deeply penetrate tumor but are easily cleared. Most of the current fixed-size nanoparticles are difficult to balance the retention and penetration, while the proposal of size-adjustable nano-drug delivery systems offers a solution to this paradox. Many endogenous and exogenous stimuli, such as acidic pH, upregulated enzymes, temperature, light, catalysts, redox conditions, and reactive oxygen species, can trigger the in situ transformation of nanoparticles based on protonation, hydrolysis, click reaction, phase transition, photoisomerization, redox reaction, etc. In this review, we summarize the principles and applications of stimuli-responsive size-adjustable strategies, including size-enlargement strategies and size-shrinkage strategies. We also propose the challenges faced by size-adjustable nano-drug delivery systems, hoping to promote the development of this strategy.
Collapse
Affiliation(s)
- Miao Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Jing-Dong Rao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Rong Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
39
|
Glassman PM, Hood ED, Ferguson LT, Zhao Z, Siegel DL, Mitragotri S, Brenner JS, Muzykantov VR. Red blood cells: The metamorphosis of a neglected carrier into the natural mothership for artificial nanocarriers. Adv Drug Deliv Rev 2021; 178:113992. [PMID: 34597748 PMCID: PMC8556370 DOI: 10.1016/j.addr.2021.113992] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/26/2021] [Accepted: 09/24/2021] [Indexed: 12/18/2022]
Abstract
Drug delivery research pursues many types of carriers including proteins and other macromolecules, natural and synthetic polymeric structures, nanocarriers of diverse compositions and cells. In particular, liposomes and lipid nanoparticles represent arguably the most advanced and popular human-made nanocarriers, already in multiple clinical applications. On the other hand, red blood cells (RBCs) represent attractive natural carriers for the vascular route, featuring at least two distinct compartments for loading pharmacological cargoes, namely inner space enclosed by the plasma membrane and the outer surface of this membrane. Historically, studies of liposomal drug delivery systems (DDS) astronomically outnumbered and surpassed the RBC-based DDS. Nevertheless, these two types of carriers have different profile of advantages and disadvantages. Recent studies showed that RBC-based drug carriers indeed may feature unique pharmacokinetic and biodistribution characteristics favorably changing benefit/risk ratio of some cargo agents. Furthermore, RBC carriage cardinally alters behavior and effect of nanocarriers in the bloodstream, so called RBC hitchhiking (RBC-HH). This article represents an attempt for the comparative analysis of liposomal vs RBC drug delivery, culminating with design of hybrid DDSs enabling mutual collaborative advantages such as RBC-HH and camouflaging nanoparticles by RBC membrane. Finally, we discuss the key current challenges faced by these and other RBC-based DDSs including the issue of potential unintended and adverse effect and contingency measures to ameliorate this and other concerns.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Laura T Ferguson
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Don L Siegel
- Department of Pathology & Laboratory Medicine, Division of Transfusion Medicine & Therapeutic Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02138, United States
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, Center for Targeted Therapeutics and Translational Nanomedicine of the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
40
|
Tereshkina YA, Torkhovskaya TI, Tikhonova EG, Kostryukova LV, Sanzhakov MA, Korotkevich EI, Khudoklinova YY, Orlova NA, Kolesanova EF. Nanoliposomes as drug delivery systems: safety concerns. J Drug Target 2021; 30:313-325. [PMID: 34668814 DOI: 10.1080/1061186x.2021.1992630] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The review highlights the safety issues of drug delivery systems based on liposomes. Due to their small sizes (about 80-120 nm, sometimes even smaller), phospholipid nanoparticles interact intensively with living systems during parenteral administration. This interaction significantly affects both their transport role and safety; therefore, special attention is paid to these issues. The review summarises the data on the basic factors affecting the safety of nanoliposomes: composition, size, surface charge, stability, the release of an incorporated drug, penetration into tissues, interaction with the complement system. Attention is paid to the authors' own research of unique phospholipid nanoparticles with a diameter of 20-30 nm. The influence of technological processes of nanoliposome production on their properties is considered. The article also discusses the modern safety assessment criteria contained in the preliminary regulatory documents of the manufacturing countries for new nanoliposome-based drugs being developed or used in the clinic.
Collapse
Affiliation(s)
- Yu A Tereshkina
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - T I Torkhovskaya
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - E G Tikhonova
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - L V Kostryukova
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - M A Sanzhakov
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - E I Korotkevich
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - Yu Yu Khudoklinova
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - N A Orlova
- Laboratory of Phospholipid Nanoparticles and Transport Systems, Institute of Biomedical Chemistry, Moscow, Russia
| | - E F Kolesanova
- Laboratory of Peptide Engineering, Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
41
|
Gheybi F, Alavizadeh SH, Rezayat SM, Hatamipour M, Akhtari J, Faridi Majidi R, Badiee A, Jaafari MR. pH-Sensitive PEGylated Liposomal Silybin: Synthesis, In Vitro and In Vivo Anti-Tumor Evaluation. J Pharm Sci 2021; 110:3919-3928. [PMID: 34418455 DOI: 10.1016/j.xphs.2021.08.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 08/14/2021] [Accepted: 08/14/2021] [Indexed: 12/24/2022]
Abstract
The drug delivery systems improve the efficacy of chemotherapeutics through enhanced targeting and controlled release however, biological barriers of tumor microenvironment greatly impede the penetration of nanomedicine within the tumor. We report herein the fabrication of a PEG-detachable silybin (SLB) pH-sensitive liposome decorated with TAT-peptide. For this, Acyl hydrazide-activated PEG2000 was prepared and linked with ketone-derivatized DPPE via an acid-labile hydrazone bond to form mPEG2000-HZ-DPPE. TAT peptide was conjugated with a shorter -PEG1000-DSPE spacer and post-inserted into PEGylated liposome (DPPC: mPEG2000-DSPE: Chol). To prepare nanoliposomes (around 100 nm), first, a novel method was used to prepare SLB-Soya PC (SLB-SPC) complex, then this complex was incorporated into nanoliposomes. The pH-sensitivity and shielding effect of long PEG chain on TAT peptide was investigated using DiI liposome and FACS analysis. Pre-treatment to the lowered pH enhanced cellular association of TAT-modified pH-sensitive liposome due to the cleavage of hydrazone bond and TAT exposure. Besides, TAT-modified pH-sensitive liposomes significantly reduced cell viability compared to the plain liposome. In vivo results were very promising with pH-sensitive liposome by detaching PEG moieties upon exposure to the acidic tumor microenvironment, enhancing cellular uptake, retarding tumor growth, and prolonging the survival of 4T1 breast tumor-bearing BALB/c mice. TAT modification of pH-sensitive liposome improved cancer cell association and cytotoxicity and demonstrated potential intracellular delivery upon exposure to acidic pH. However, in in vivo studies, TAT as a targeting ligand significantly decreased the therapeutic efficacy of the formulation attributed to an inefficient tumor accumulation and higher release rate in the circulation. The results of this study indicated that pH-sensitive liposome containing SLB, which was prepared with a novel method with a significant SLB loading efficiency, is very effective in the treatment of 4T1 breast tumor-bearing BALB/c mice and merits further investigation.
Collapse
Affiliation(s)
- Fatemeh Gheybi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Hatamipour
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Akhtari
- Department of Medical Nanotechnology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Faridi Majidi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
42
|
Abbasi Kajani A, Haghjooy Javanmard S, Asadnia M, Razmjou A. Recent Advances in Nanomaterials Development for Nanomedicine and Cancer. ACS APPLIED BIO MATERIALS 2021; 4:5908-5925. [PMID: 35006909 DOI: 10.1021/acsabm.1c00591] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer is considered one of the leading causes of death, with a growing number of cases worldwide. However, the early diagnosis and efficient therapy of cancer have remained a critical challenge. The emergence of nanomedicine has opened up a promising window to address the drawbacks of cancer detection and treatment. A wide range of engineered nanomaterials and nanoplatforms with different shapes, sizes, and composition has been developed for various biomedical applications. Nanomaterials have been increasingly used in various applications in bioimaging, diagnosis, and therapy of cancers. Recently, numerous multifunctional and smart nanoparticles with the ability of simultaneous diagnosis and targeted cancer therapy have been reported. The multidisciplinary attempts led to the development of several exciting clinically approved nanotherapeutics. The nanobased materials and devices have also been used extensively to develop point-of-care and highly sensitive methods of cancer detection. In this review article, the most significant achievements and latest advances in the nanomaterials development for cancer nanomedicine are critically discussed. In addition, the future perspectives of this field are evaluated.
Collapse
Affiliation(s)
- Abolghasem Abbasi Kajani
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Mohsen Asadnia
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Amir Razmjou
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 73441-81746, Iran
- UNESCO Centre for Membrane Science and Technology, School of Chemical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
- Centre for Technology in Water and Wastewater, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| |
Collapse
|
43
|
Zhuang Y, Zhao Y, Wang B, Wang Q, Cai T, Cai Y. Strategies for Preparing Different Types of Lipid Polymer Hybrid Nanoparticles in Targeted Tumor Therapy. Curr Pharm Des 2021; 27:2274-2288. [PMID: 33222665 DOI: 10.2174/1381612826666201120155558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/27/2020] [Indexed: 11/22/2022]
Abstract
At present, cancer is one of the most common diseases in the world, causing a large number of deaths and seriously affecting people's health. The traditional treatment of cancer is mainly surgery, radiotherapy or chemotherapy. Conventional chemotherapy is still an important treatment, but it has some shortcomings, such as poor cell selectivity, serious side effects, drug resistance and so on. Nanoparticle administration can improve drug stability, reduce toxicity, prolong drug release time, prolong system half-life, and bring broad prospects for tumor therapy. Lipid polymer hybrid nanoparticles (LPNs), which combine the advantages of polymer core and phospholipid shell to form a single platform, have become multi-functional drug delivery platforms. This review introduces the basic characteristics, structure and preparation methods of LPNs, and discusses targeting strategies of LPNs in tumor therapy in order to overcome the defects of traditional drug therapy.
Collapse
Affiliation(s)
- Yong Zhuang
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yiye Zhao
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Bingyue Wang
- Guangzhou Jiayuan Medical and Pharmaceutical Technology Co., Ltd., Guangzhou 510663, China
| | - Qi Wang
- Guangzhou Jiayuan Medical and Pharmaceutical Technology Co., Ltd., Guangzhou 510663, China
| | - Tiange Cai
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| |
Collapse
|
44
|
Baker A, Khan MS, Iqbal MZ, Khan MS. Tumor-targeted Drug Delivery by Nanocomposites. Curr Drug Metab 2021; 21:599-613. [PMID: 32433002 DOI: 10.2174/1389200221666200520092333] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 01/30/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Tumor-targeted delivery by nanoparticles is a great achievement towards the use of highly effective drug at very low doses. The conventional development of tumor-targeted delivery by nanoparticles is based on enhanced permeability and retention (EPR) effect and endocytosis based on receptor-mediated are very demanding due to the biological and natural complications of tumors as well as the restrictions on the design of the accurate nanoparticle delivery systems. METHODS Different tumor environment stimuli are responsible for triggered multistage drug delivery systems (MSDDS) for tumor therapy and imaging. Physicochemical properties, such as size, hydrophobicity and potential transform by MSDDS because of the physiological blood circulation different, intracellular tumor environment. This system accomplishes tumor penetration, cellular uptake improved, discharge of drugs on accurate time, and endosomal discharge. RESULTS Maximum drug delivery by MSDDS mechanism to target therapeutic cells and also tumor tissues and sub cellular organism. Poorly soluble compounds and bioavailability issues have been faced by pharmaceutical industries, which are resolved by nanoparticle formulation. CONCLUSION In our review, we illustrate different types of triggered moods and stimuli of the tumor environment, which help in smart multistage drug delivery systems by nanoparticles, basically a multi-stimuli sensitive delivery system, and elaborate their function, effects, and diagnosis.
Collapse
Affiliation(s)
- Abu Baker
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Mohd Salman Khan
- Clinical Biochemistry & Natural Product Research Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Muhammad Zafar Iqbal
- Department of Studies and Research in Zoology, Government First Grade College, Karwar, 581301, India
| | - Mohd Sajid Khan
- Nanomedicine & Nanobiotechnology Lab, Department of Biosciences, Integral University, Lucknow, 226026, India
| |
Collapse
|
45
|
Khan S, Vahdani Y, Hussain A, Haghighat S, Heidari F, Nouri M, Haj Bloukh S, Edis Z, Mahdi Nejadi Babadaei M, Ale-Ebrahim M, Hasan A, Sharifi M, Bai Q, Hassan M, Falahati M. Polymeric micelles functionalized with cell penetrating peptides as potential pH-sensitive platforms in drug delivery for cancer therapy: A review. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
46
|
He B, Sui X, Yu B, Wang S, Shen Y, Cong H. Recent advances in drug delivery systems for enhancing drug penetration into tumors. Drug Deliv 2021; 27:1474-1490. [PMID: 33100061 PMCID: PMC7594734 DOI: 10.1080/10717544.2020.1831106] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The emergence of nanomaterials for drug delivery provides the opportunity to avoid the side effects of systemic drug administration and injury caused by the removal of tumors, delivering great promise for future cancer treatments. However, the efficacy of current nano drugs is not significantly better than that of the original drug treatments. The important reason is that nano drugs enter the tumor vasculature, remaining close to the blood vessels and unable to enter the tumor tissue or tumor cells to complete the drug delivery process. The low efficiency of drug penetration into tumors has become a bottleneck restricting the development of nano-drugs. Herein, we present a systematic overview of recent advances on the design of nano-drug carriers in drug delivery systems for enhancing drug penetration into tumors. The review is organized into four sections: The drug penetration process in tumor tissue includes paracellular and transcellular transport, which is summarized first. Strategies that promote tumor penetration are then introduced, including methods of remodeling the tumor microenvironment, charge inversion, dimensional change, and surface modification of ligands which promote tissue penetration. Conclusion and the prospects for the future development of drug penetration are finally briefly illustrated. The review is intended to provide thoughts for effective treatment of cancer by summarizing strategies for promoting the endocytosis of nano drugs into tumor cells.
Collapse
Affiliation(s)
- Bin He
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xin Sui
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Key Laboratory of Bio-Fibers and Eco-Textiles, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, China
| | - Song Wang
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Key Laboratory of Bio-Fibers and Eco-Textiles, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, China
| |
Collapse
|
47
|
Li H, Wang Y, Tang Q, Yin D, Tang C, He E, Zou L, Peng Q. The protein corona and its effects on nanoparticle-based drug delivery systems. Acta Biomater 2021; 129:57-72. [PMID: 34048973 DOI: 10.1016/j.actbio.2021.05.019] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 05/18/2021] [Indexed: 02/04/2023]
Abstract
In most cases, once nanoparticles (NPs) enter the blood, their surface is covered by biological molecules, especially proteins, forming a so-called protein corona (PC). As a result, what the cells of the body "see" is not the NPs as formulated by the chemists, but the PC. In this way, the PC can influence the effects of the NPs and even mask the desired effects of the NP components. While this can argue for trying to inhibit protein-nanomaterial interactions, encapsulating NPs in an endogenous PC may increase their clinical usefulness. In this review, we briefly introduce the concept of the PC, its formation and its effects on the behavior of NPs. We also discuss how to reduce the formation of PCs or exploit them to enhance NP functions. Studying the interactions between proteins and NPs will provide insights into their clinical activity in health and disease. STATEMENT OF SIGNIFICANCE: The formation of protein corona (PC) will affect the operation of nanoparticles (NPs) in vivo. Since there are many proteins in the blood, it is impossible to completely overcome the formation of PC. Therefore, the use of PCs to deliver drug is the best choice. De-opsonins adsorbed on NPs can reduce macrophage phagocytosis and cytotoxicity of NPs, and prolong their circulation in blood. Albumin, apolipoprotein and transferrin are typical de-opsonins. In present review, we mainly discuss how to optimize the delivery of nanoparticles through the formation of albumin corona, transferrin corona and apolipoprotein corona in vivo or in vitro.
Collapse
Affiliation(s)
- Hanmei Li
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China
| | - Yao Wang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Qi Tang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Dan Yin
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Chuane Tang
- School of Mechanical Engineering, Chengdu university, Chengdu 610106, China
| | - En He
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu university, Chengdu 610106, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
48
|
Huang S, Ren D, Wu X, Li M, Yu X, Nie X, Wang Y, Wang Y. Glycyrrhetinic Acid and TAT Peptide Modified Dual-functional Liposomes for Treatment of Hepatocellular Cancer. Curr Top Med Chem 2021; 20:2493-2505. [PMID: 32703132 DOI: 10.2174/1568026620666200722110244] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 02/15/2020] [Accepted: 03/10/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Surgery remains the front-line therapeutic strategy to treat early hepatocellular carcinoma (HCC). However, the 5-year recurrence rates of HCC patients are high. 10- Hydroxycamptothecin (10-HCPT) is a known anti-HCC agent but its poor solubility and bioavailability have limited its clinical use. OBJECTIVE In this study, we developed a novel nanoliposome encapsulated 10-hydroxycamptothecin modified with glycyrrhetinic acid (GA) and TAT peptide (GA/TAT-HCPT-LP) for the treatment of HCC. Dual modified GA and TAT can enhance tumor targeting and tumor penetration. METHODS The GA/TAT-HCPT-LP NPs were synthesized using the thin-film dispersion method. GA/TAT-HCPT-LP were characterized for particle size, zeta potential and morphology. Drug release from the GA/TAT-HCPT-LP liposomes was measured by dialysis. Cell-uptake was assessed by microscopy and flow cytometry. Cell proliferation, migration and apoptosis were measured to evaluate in vitro antitumor activity of GA/TAT-HCPT-LP via CCK-8 assays, Transwell assays, and flow cytometry, respectively. The in vivo distribution of GA/TAT-HCPT-LP was evaluated in HCC animal models. Tumor- bearing mouse models were used to assess the in vivo therapeutic efficacy of GA/TAT-HCPT-LP. RESULTS The mean particle size and mean zeta potential of GA/TAT-HCPT-LP were 135.55 ± 2.76 nm and -4.57 ± 0.23 mV, respectively. Transmission electron micrographs (TEM) showed that the GA/TAT-HCPT-LP had a near spherical shape and a double-membrane structure. GA/TAT-HCPT-LP led to slow and continuous drug release, and could bind to HepG2 cells more readily than other groups. Compared to control groups, treatment with GA/TAT-HCPT-LP had a significantly large effect on inhibiting cell proliferation, tumor cell migration and cell apoptosis. In vivo assays showed that GA/TATHCPT- LP selectively accumulated in tumor tissue with obvious antitumor efficacy. CONCLUSION In conclusion, the synthesized GA/TAT-HCPT-LP could effectively target tumor cells and enhance cell penetration, highlighting its potential for hepatocellular cancer therapy.
Collapse
Affiliation(s)
- Sixi Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006 Guangzhou, China
| | - Di Ren
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006 Guangzhou, China
| | - Xinrong Wu
- General Hospital of Southern Theater Command, PLA, 510006 Guangzhou, China
| | - Ming Li
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, China
| | - Xuesong Yu
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, China
| | - Xiaoling Nie
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006 Guangzhou, China
| | - Ying Wang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, 510006 Guangzhou, China
| | - Yan Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, 510006 Guangzhou, China
| |
Collapse
|
49
|
Ashique S, Sandhu NK, Chawla V, Chawla PA. Targeted Drug Delivery: Trends and Perspectives. Curr Drug Deliv 2021; 18:1435-1455. [PMID: 34151759 DOI: 10.2174/1567201818666210609161301] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/03/2021] [Accepted: 04/07/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Due to various limitations in conventional drug delivery system, it is important to focus on the target-specific drug delivery system where we can deliver the drug without any degradation. Among various challenges faced by a formulation scientist, delivering the drug to its right site, in its right dose, is also an important aim. A focused drug transport aims to extend, localize, target and have a safe drug interaction with the diseased tissue. OBJECTIVE The aim of targeted drug delivery is to make the required amount of the drug available at its desired site of action. Drug targeting can be accomplished in a number ways that include enzyme mediation, pH-dependent release, use of special vehicles, receptor targeting among other mechanisms. Intelligently designed targeted drug delivery systems also offer the advantages of a low dose of the drug along with reduced side effects which ultimately improves patient compliance. Incidences of dose dumping and dosage form failure are negligible. A focused drug transport aims to have a safe drug interaction with the diseased tissue. CONCLUSION This review focuses on the available targeting techniques for delivery to the colon, brain and other sites of interest. Overall, the article should make an excellent read for the researchers in this area. Newer drug targets may be identified and exploited for successful drug targeting.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Navjot Kaur Sandhu
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Viney Chawla
- University Institute of Pharmaceutical Sciences and Research, Baba Farid University of Health Sciences, Faridkot, Punjab, India
| | - Pooja A Chawla
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|
50
|
Evaluation of Nanotargeted 111In-Cyclic RGDfK-Liposome in a Human Melanoma Xenotransplantation Model. Int J Mol Sci 2021; 22:ijms22031099. [PMID: 33499267 PMCID: PMC7866009 DOI: 10.3390/ijms22031099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Nanotargeted liposomes may be modified with targeting peptide on the surface of a prepared liposome to endow specificity and elevate targeting efficiency. The aim of this study was to develop a radioactive targeted nanoparticle, the 111In-cyclic RGDfK-liposome, and its advantage of recognizing the αVβ3 integrin was examined. The cyclic RGDfK modified liposomes were demonstrated the ability to bind the αVβ3 integrin expressed on the surface of human melanoma cell in vitro and in vivo. The effects of the cyclic RGDfK-liposome on the functioning of phagocytes was also examined, showing no considerable negative effects on the engulfment of bacteria and the generation of reactive oxygen species. Based upon these findings, the cyclic RGDfK- liposome is said to be a promising agent for tumor imaging.
Collapse
|