1
|
Eivazzadeh-Keihan R, Saadatidizaji Z, Mahdavi M, Maleki A, Irani M, Zare I. Recent advances in gold nanoparticles-based biosensors for tuberculosis determination. Talanta 2024; 275:126099. [PMID: 38640517 DOI: 10.1016/j.talanta.2024.126099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/16/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
Tuberculosis (TB) is one of the major killer diseases affecting lung parenchymal tissues. Mycobacterium tuberculosis (Mtb) is the bacterium that causes it. It most commonly affects the lungs, although it can affect any part of the body, including the stomach, glands, bones, and nervous system. Although anti-mycobacterial drugs are available, it remains a major threat to public health due to the rise of drug-resistant strains, and early and accurate diagnosis is very important. Currently, research science and medical communities are focusing on the use of cost-effective biosensors to manage human biological processes and assess accurate health diagnostics. Due to their high sensitivity in chemical and biological assays, nanomaterials have been considered in the field of biosensors for better diagnosis, and among them, gold nanoparticles (AuNPs) can play an important role in accelerating the diagnosis of TB. Superior biocompatibility, conductivity, catalytic properties, high surface-to-volume ratio, and high density enable their widespread use in the fabrication of biosensors. This review evaluates the diagnostic accuracy of AuNP-based biosensors for the detection of Mtb. According to different transducers of biosensors, their structure, performance, advantages and limitations are summarized and compared. Moreover, the upcoming challenges in their analytical performance have been highlighted and the strategies to overcome those challenges have been briefly discussed.
Collapse
Affiliation(s)
- Reza Eivazzadeh-Keihan
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran.
| | - Zahra Saadatidizaji
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Maleki
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran.
| | - Mohammad Irani
- Department of Pharmaceutics, School of Pharmacy, Alborz University of Medical Sciences, Karaj, Iran
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co., Ltd., Shiraz, 7178795844, Iran.
| |
Collapse
|
2
|
Yan X, Chen Q. Polyamidoamine Dendrimers: Brain-Targeted Drug Delivery Systems in Glioma Therapy. Polymers (Basel) 2024; 16:2022. [PMID: 39065339 PMCID: PMC11280609 DOI: 10.3390/polym16142022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Glioma is the most common primary intracranial tumor, which is formed by the malignant transformation of glial cells in the brain and spinal cord. It has the characteristics of high incidence, high recurrence rate, high mortality and low cure rate. The treatments for glioma include surgical removal, chemotherapy and radiotherapy. Due to the obstruction of the biological barrier of brain tissue, it is difficult to achieve the desired therapeutic effects. To address the limitations imposed by the brain's natural barriers and enhance the treatment efficacy, researchers have effectively used brain-targeted drug delivery systems (DDSs) in glioma therapy. Polyamidoamine (PAMAM) dendrimers, as branched macromolecular architectures, represent promising candidates for studies in glioma therapy. This review focuses on PAMAM-based DDSs in the treatment of glioma, highlighting their physicochemical characteristics, structural properties as well as an overview of the toxicity and safety profiles.
Collapse
Affiliation(s)
- Xinyi Yan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Qi Chen
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
3
|
Nakamura N, Ohta S. Precise control methods of the physicochemical properties of nanoparticles for personalized medicine. Curr Opin Biotechnol 2024; 87:103108. [PMID: 38513338 DOI: 10.1016/j.copbio.2024.103108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 02/25/2024] [Accepted: 02/25/2024] [Indexed: 03/23/2024]
Abstract
Biomedical applications of nanoparticles (NPs) have attracted much attention. With the advancement of personalized medicine, researchers are now proposing the concept that the design of NPs needs to be optimized according to the individual patient. To realize this concept, an important question is how precisely we can tailor the physicochemical properties of NPs, such as size, shape, and surface chemistry, using current technology. This review discusses recent advances and challenges in the precise control of the size, shape, and surface chemistry of NPs. While control methods have advanced significantly over the past 20 years, the size, shape, and surface chemistry of currently available NPs vary by type, requiring careful selection based on the targeted disease, organ, and patient.
Collapse
Affiliation(s)
- Noriko Nakamura
- Institute of Engineering Innovation, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo 113-8656, Japan; Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Seiichi Ohta
- Institute of Engineering Innovation, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo 113-8656, Japan; Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; Department of Chemical System Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
4
|
Lei T, Yang Z, Li H, Qin M, Gao H. Interactions between nanoparticles and pathological changes of vascular in Alzheimer's disease. Adv Drug Deliv Rev 2024; 207:115219. [PMID: 38401847 DOI: 10.1016/j.addr.2024.115219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
Emerging evidence suggests that vascular pathological changes play a pivotal role in the pathogenesis of Alzheimer's disease (AD). The dysfunction of the cerebral vasculature occurs in the early course of AD, characterized by alterations in vascular morphology, diminished cerebral blood flow (CBF), impairment of the neurovascular unit (NVU), vasculature inflammation, and cerebral amyloid angiopathy. Vascular dysfunction not only facilitates the influx of neurotoxic substances into the brain, triggering inflammation and immune responses but also hampers the efflux of toxic proteins such as Aβ from the brain, thereby contributing to neurodegenerative changes in AD. Furthermore, these vascular changes significantly impact drug delivery and distribution within the brain. Therefore, developing targeted delivery systems or therapeutic strategies based on vascular alterations may potentially represent a novel breakthrough in AD treatment. This review comprehensively examines various aspects of vascular alterations in AD and outlines the current interactions between nanoparticles and pathological changes of vascular.
Collapse
Affiliation(s)
- Ting Lei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zixiao Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanmei Li
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Meng Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
5
|
Wang J, Wang Z, Zhang G, Rodrigues J, Tomás H, Shi X, Shen M. Blood-brain barrier-crossing dendrimers for glioma theranostics. Biomater Sci 2024; 12:1346-1356. [PMID: 38362780 DOI: 10.1039/d4bm00043a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Glioma, as a disease of the central nervous system, is difficult to be treated due to the presence of the blood-brain barrier (BBB) that can severely hamper the efficacy of most therapeutic agents. Hence, drug delivery to glioma in an efficient, safe, and specifically targeted manner is the key to effective treatment of glioma. With the advances in nanotechnology, targeted drug delivery systems have been extensively explored to deliver chemotherapeutic agents, nucleic acids, and contrast agents. Among these nanocarriers, dendrimers have played a significant role since they possess highly branched structures, and are easy to be decorated, thus offering numerous binding sites for various drugs and ligands. Dendrimers can be designed to cross the BBB for glioma targeting, therapy or theranostics. In this review, we provide a concise overview of dendrimer-based carrier designs including dendrimer surface modification with hydroxyl termini, peptides, and transferrin etc. for glioma imaging diagnostics, chemotherapy, gene therapy, or imaging-guided therapy. Finally, the future perspectives of dendrimer-based glioma theraputics are also briefly discussed.
Collapse
Affiliation(s)
- Jinxia Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Zhiqiang Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Guixiang Zhang
- Department of Radiology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
| | - João Rodrigues
- CQM-Centro de Quimica da Madeira, Universidade da Madeira, Funchal 9020-105, Portugal
| | - Helena Tomás
- CQM-Centro de Quimica da Madeira, Universidade da Madeira, Funchal 9020-105, Portugal
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
- CQM-Centro de Quimica da Madeira, Universidade da Madeira, Funchal 9020-105, Portugal
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
6
|
Bhunia S, Kolishetti N, Vashist A, Yndart Arias A, Brooks D, Nair M. Drug Delivery to the Brain: Recent Advances and Unmet Challenges. Pharmaceutics 2023; 15:2658. [PMID: 38139999 PMCID: PMC10747851 DOI: 10.3390/pharmaceutics15122658] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/02/2023] [Accepted: 11/08/2023] [Indexed: 12/24/2023] Open
Abstract
Brain cancers and neurodegenerative diseases are on the rise, treatments for central nervous system (CNS) diseases remain limited. Despite the significant advancement in drug development technology with emerging biopharmaceuticals like gene therapy or recombinant protein, the clinical translational rate of such biopharmaceuticals to treat CNS disease is extremely poor. The blood-brain barrier (BBB), which separates the brain from blood and protects the CNS microenvironment to maintain essential neuronal functions, poses the greatest challenge for CNS drug delivery. Many strategies have been developed over the years which include local disruption of BBB via physical and chemical methods, and drug transport across BBB via transcytosis by targeting some endogenous proteins expressed on brain-capillary. Drug delivery to brain is an ever-evolving topic, although there were multiple review articles in literature, an update is warranted due to continued growth and new innovations of research on this topic. Thus, this review is an attempt to highlight the recent strategies employed to overcome challenges of CNS drug delivery while emphasizing the necessity of investing more efforts in CNS drug delivery technologies parallel to drug development.
Collapse
Affiliation(s)
- Sukanya Bhunia
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Nagesh Kolishetti
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Arti Vashist
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Adriana Yndart Arias
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Deborah Brooks
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
7
|
Babu B, Stoltz SA, Mittal A, Pawar S, Kolanthai E, Coathup M, Seal S. Inorganic Nanoparticles as Radiosensitizers for Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2873. [PMID: 37947718 PMCID: PMC10647410 DOI: 10.3390/nano13212873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Nanotechnology has expanded what can be achieved in our approach to cancer treatment. The ability to produce and engineer functional nanoparticle formulations to elicit higher incidences of tumor cell radiolysis has resulted in substantial improvements in cancer cell eradication while also permitting multi-modal biomedical functionalities. These radiosensitive nanomaterials utilize material characteristics, such as radio-blocking/absorbing high-Z atomic number elements, to mediate localized effects from therapeutic irradiation. These materials thereby allow subsequent scattered or emitted radiation to produce direct (e.g., damage to genetic materials) or indirect (e.g., protein oxidation, reactive oxygen species formation) damage to tumor cells. Using nanomaterials that activate under certain physiologic conditions, such as the tumor microenvironment, can selectively target tumor cells. These characteristics, combined with biological interactions that can target the tumor environment, allow for localized radio-sensitization while mitigating damage to healthy cells. This review explores the various nanomaterial formulations utilized in cancer radiosensitivity research. Emphasis on inorganic nanomaterials showcases the specific material characteristics that enable higher incidences of radiation while ensuring localized cancer targeting based on tumor microenvironment activation. The aim of this review is to guide future research in cancer radiosensitization using nanomaterial formulations and to detail common approaches to its treatment, as well as their relations to commonly implemented radiotherapy techniques.
Collapse
Affiliation(s)
- Balaashwin Babu
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
| | - Samantha Archer Stoltz
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Agastya Mittal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Shreya Pawar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
| | - Melanie Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA;
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Nanoscience Technology Center, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
8
|
Chen C, Cai QW, Zhan CZ, Wang BC, Li PF, Xie R, Ju XJ, Liu Z, Wang W, Chu LY. Controllable Fabrication of Highly Uniform Sub-10 nm Nanoparticles from Spontaneous Confined Nanoemulsification. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300801. [PMID: 37072877 DOI: 10.1002/smll.202300801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/30/2023] [Indexed: 05/03/2023]
Abstract
Sub-10 nm nanoparticles are known to exhibit extraordinary size-dependent properties for wide applications. Many approaches have been developed for synthesizing sub-10 nm inorganic nanoparticles, but the fabrication of sub-10 nm polymeric nanoparticles is still challenging. Here, a scalable, spontaneous confined nanoemulsification strategy that produces uniform sub-10 nm nanodroplets for template synthesis of sub-10 nm polymeric nanoparticles is proposed. This strategy introduces a high-concentration interfacial reaction to create overpopulated surfactants that are insoluble at the droplet surface. These overpopulated surfactants act as barriers, resulting in highly accumulated surfactants inside the droplet via a confined reaction. These surfactants exhibit significantly changed packing geometry, solubility, and interfacial activity to enhance the molecular-level impact on interfacial instability for creating sub-10 nm nanoemulsions via self-burst nanoemulsification. Using the nanodroplets as templates, the fabrication of uniform sub-10 nm polymeric nanoparticles, as small as 3.5 nm, made from biocompatible polymers and capable of efficient drug encapsulation is demonstrated. This work opens up brand-new opportunities to easily create sub-10 nm nanoemulsions and advanced ultrasmall functional nanoparticles.
Collapse
Affiliation(s)
- Chen Chen
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- Department of Chemical Engineering, Shanghai Jiaotong University, Shanghai, 200240, China
| | - Quan-Wei Cai
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Cai-Zhen Zhan
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Bi-Cong Wang
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Ping-Fan Li
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Rui Xie
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xiao-Jie Ju
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Zhuang Liu
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Wei Wang
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Liang-Yin Chu
- School of Chemical Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, Sichuan, 610065, China
| |
Collapse
|
9
|
Boltman T, Meyer M, Ekpo O. Diagnostic and Therapeutic Approaches for Glioblastoma and Neuroblastoma Cancers Using Chlorotoxin Nanoparticles. Cancers (Basel) 2023; 15:3388. [PMID: 37444498 DOI: 10.3390/cancers15133388] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 07/15/2023] Open
Abstract
Glioblastoma multiforme (GB) and high-risk neuroblastoma (NB) are known to have poor therapeutic outcomes. As for most cancers, chemotherapy and radiotherapy are the current mainstay treatments for GB and NB. However, the known limitations of systemic toxicity, drug resistance, poor targeted delivery, and inability to access the blood-brain barrier (BBB), make these treatments less satisfactory. Other treatment options have been investigated in many studies in the literature, especially nutraceutical and naturopathic products, most of which have also been reported to be poorly effective against these cancer types. This necessitates the development of treatment strategies with the potential to cross the BBB and specifically target cancer cells. Compounds that target the endopeptidase, matrix metalloproteinase 2 (MMP-2), have been reported to offer therapeutic insights for GB and NB since MMP-2 is known to be over-expressed in these cancers and plays significant roles in such physiological processes as angiogenesis, metastasis, and cellular invasion. Chlorotoxin (CTX) is a promising 36-amino acid peptide isolated from the venom of the deathstalker scorpion, Leiurus quinquestriatus, demonstrating high selectivity and binding affinity to a broad-spectrum of cancers, especially GB and NB through specific molecular targets, including MMP-2. The favorable characteristics of nanoparticles (NPs) such as their small sizes, large surface area for active targeting, BBB permeability, etc. make CTX-functionalized NPs (CTX-NPs) promising diagnostic and therapeutic applications for addressing the many challenges associated with these cancers. CTX-NPs may function by improving diffusion through the BBB, enabling increased localization of chemotherapeutic and genotherapeutic drugs to diseased cells specifically, enhancing imaging modalities such as magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), optical imaging techniques, image-guided surgery, as well as improving the sensitization of radio-resistant cells to radiotherapy treatment. This review discusses the characteristics of GB and NB cancers, related treatment challenges as well as the potential of CTX and its functionalized NP formulations as targeting systems for diagnostic, therapeutic, and theranostic purposes. It also provides insights into the potential mechanisms through which CTX crosses the BBB to bind cancer cells and provides suggestions for the development and application of novel CTX-based formulations for the diagnosis and treatment of GB and NB in the future.
Collapse
Affiliation(s)
- Taahirah Boltman
- Department of Medical Biosciences, University of the Western Cape, Robert Sobukwe Road, Bellville, Cape Town 7535, South Africa
| | - Mervin Meyer
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, University of the Western Cape, Robert Sobukwe Road, Bellville, Cape Town 7535, South Africa
| | - Okobi Ekpo
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
10
|
Vanbilloen WJF, Rechberger JS, Anderson JB, Nonnenbroich LF, Zhang L, Daniels DJ. Nanoparticle Strategies to Improve the Delivery of Anticancer Drugs across the Blood-Brain Barrier to Treat Brain Tumors. Pharmaceutics 2023; 15:1804. [PMID: 37513992 PMCID: PMC10383584 DOI: 10.3390/pharmaceutics15071804] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Primary brain and central nervous system (CNS) tumors are a diverse group of neoplasms that occur within the brain and spinal cord. Although significant advances in our understanding of the intricate biological underpinnings of CNS neoplasm tumorigenesis and progression have been made, the translation of these discoveries into effective therapies has been stymied by the unique challenges presented by these tumors' exquisitely sensitive location and the body's own defense mechanisms (e.g., the brain-CSF barrier and blood-brain barrier), which normally protect the CNS from toxic insult. These barriers effectively prevent the delivery of therapeutics to the site of disease. To overcome these obstacles, new methods for therapeutic delivery are being developed, with one such approach being the utilization of nanoparticles. Here, we will cover the current state of the field with a particular focus on the challenges posed by the BBB, the different nanoparticle classes which are under development for targeted CNS tumor therapeutics delivery, and strategies which have been developed to bypass the BBB and enable effective therapeutics delivery to the site of disease.
Collapse
Affiliation(s)
- Wouter J. F. Vanbilloen
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Neurology, Elisabeth-Tweesteden Hospital, 5022 GC Tilburg, The Netherlands
| | - Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jacob B. Anderson
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
- Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Leo F. Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Hopp Children’s Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - Liang Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
11
|
Zhao F, Zhang X, Bai F, Lei S, He G, Huang P, Lin J. Maximum Emission Peak Over 1500 nm of Organic Assembly for Blood-Brain Barrier-Crossing NIR-IIb Phototheranostics of Orthotopic Glioblastoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208097. [PMID: 36893436 DOI: 10.1002/adma.202208097] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/11/2023] [Indexed: 06/02/2023]
Abstract
The development of blood-brain barrier (BBB)-crossing phototheranostic agents in second near-infrared window (NIR-II), especially in the range of 1500-1700 nm (NIR-IIb), affords great opportunities for glioblastoma (GBM) management. Herein, an organic assembly (denoted as LET-12) with the maximum absorption peak at 1400 nm and emission peak at 1512 nm with trailing over 1700 nm through the self-assembly of organic small molecule IR-1064 is designed and subsequently decorated with choline and acetylcholine analogs. The LET-12 can effectively cross BBB through the brain's choline-like receptors-mediated transcytosis and accumulated in tumor tissues, thus achieving fluorescence/photoacoustic (FL/PA) duplex imaging of orthotopic GBM with ≈3.0 mm depth and a superior tumor-to-normal tissue signal ratio (20.93 ± 0.59 for FL imaging and 32.63 ± 1.16 for PA imaging, respectively). Owing to its good photothermal conversion ability, the LET-12 also can serve as a photothermal conversion agent, achieving obvious tumor repression of orthotopic murine GBM model after once treatment. The findings indicate that the LET-12 holds great potential for BBB-crossing NIR-IIb phototheranostics of orthotopic GBM. This self-assembly strategy of organic small molecules opens a new avenue for the construction of NIR-IIb phototheranostics.
Collapse
Affiliation(s)
- Feng Zhao
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Xinming Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Fang Bai
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Shan Lei
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Gang He
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| |
Collapse
|
12
|
Qiao R, Fu C, Forgham H, Javed I, Huang X, Zhu J, Whittaker AK, Davis TP. Magnetic Iron Oxide Nanoparticles for Brain Imaging and Drug Delivery. Adv Drug Deliv Rev 2023; 197:114822. [PMID: 37086918 DOI: 10.1016/j.addr.2023.114822] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/14/2023] [Accepted: 04/09/2023] [Indexed: 04/24/2023]
Abstract
Central nervous system (CNS) disorders affect as many as 1.5 billion people globally. The limited delivery of most imaging and therapeutic agents into the brain is a major challenge for treatment of CNS disorders. With the advent of nanotechnologies, controlled delivery of drugs with nanoparticles holds great promise in CNS disorders for overcoming the blood-brain barrier (BBB) and improving delivery efficacy. In recent years, magnetic iron oxide nanoparticles (MIONPs) have stood out as a promising theranostic nanoplatform for brain imaging and drug delivery as they possess unique physical properties and biodegradable characteristics. In this review, we summarize the recent advances in MIONP-based platforms as imaging and drug delivery agents for brain diseases. We firstly introduce the methods of synthesis and surface functionalization of MIONPs with emphasis on the inclusion of biocompatible polymers that allow for the addition of tailored physicochemical properties. We then discuss the recent advances in in vivo imaging and drug delivery applications using MIONPs. Finally, we present a perspective on the remaining challenges and possible future directions for MIONP-based brain delivery systems.
Collapse
Affiliation(s)
- Ruirui Qiao
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Changkui Fu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Helen Forgham
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ibrahim Javed
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Xumin Huang
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jiayuan Zhu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andrew K Whittaker
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Thomas P Davis
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
13
|
Wu JR, Hernandez Y, Miyasaki KF, Kwon EJ. Engineered nanomaterials that exploit blood-brain barrier dysfunction fordelivery to the brain. Adv Drug Deliv Rev 2023; 197:114820. [PMID: 37054953 DOI: 10.1016/j.addr.2023.114820] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023]
Abstract
The blood-brain barrier (BBB) is a highly regulated physical and functional boundarythat tightly controls the transport of materials between the blood and the brain. There is an increasing recognition that the BBB is dysfunctional in a wide range of neurological disorders; this dysfunction can be symptomatic of the disease but can also play a role in disease etiology. BBB dysfunction can be exploited for the delivery of therapeutic nanomaterials. Forexample, there can be a transient, physical disruption of the BBB in diseases such as brain injury and stroke, which allows temporary access of nanomaterials into the brain. Physicaldisruption of the BBB through external energy sources is now being clinically pursued toincrease therapeutic delivery into the brain. In other diseases, the BBB takes on new properties that can beleveraged by delivery carriers. For instance, neuroinflammation induces the expression ofreceptors on the BBB that can be targeted by ligand-modified nanomaterials and theendogenous homing of immune cells into the diseased brain can be hijacked for the delivery ofnanomaterials. Lastly, BBB transport pathways can be altered to increase nanomaterial transport. In this review, we will describe changes that can occur in the BBB in disease, and how these changes have been exploited by engineered nanomaterials forincreased transport into the brain.
Collapse
Affiliation(s)
- Jason R Wu
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Yazmin Hernandez
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Katelyn F Miyasaki
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Ester J Kwon
- Department of Bioengineering, University of California San Diego, La Jolla, CA; Sanford Consortium for Regenerative Medicine.
| |
Collapse
|
14
|
Singh S, Jaiswal V, Singh JK, Semwal R, Raina D. Nanoparticle formulations: A smart era of advanced treatment with nanotoxicological imprints on the human body. Chem Biol Interact 2023; 373:110355. [PMID: 36682480 DOI: 10.1016/j.cbi.2023.110355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/21/2022] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
In the modern era, nanoparticles are the preferred dosage form, and maximum research is going on in the field of nanoparticle formulations. But as they are so small, nanoparticles are able to slip through the body's defenses and cause damage to the organs and tissues deep inside. In recent years, most researchers have focused solely on the therapeutic value of drugs or, at times, the performance of dosage forms, but few have given toxicity studies equal weight in their research. This review demonstrates that nanoparticle formulations are not suitable from a safety standpoint. So, researchers should be focused on alternative formulations like nanoemulsion, nanogel, and other liquids as well as semisolid formulations.
Collapse
Affiliation(s)
- Siddharth Singh
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun, 248007, India
| | - Vishakha Jaiswal
- Faculty of Pharmacy, BBDNIIT, Lucknow, Uttar Pradesh, 226028, India
| | | | - Ravindra Semwal
- Research and Development Centre, Faculty of Biomedical Sciences, Uttarakhand Ayurved University, Harrawala, Dehradun, 248001, India
| | - Deepika Raina
- School of Pharmacy, Graphic Era Hill University, Dehradun, India.
| |
Collapse
|
15
|
Angolkar M, Paramshetti S, Halagali P, Jain V, Patil AB, Somanna P. Nanotechnological advancements in the brain tumor therapy: a novel approach. Ther Deliv 2023; 13:531-557. [PMID: 36802944 DOI: 10.4155/tde-2022-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Nanotechnological advancements over the past few years have led to the development of newer treatment strategies in brain cancer therapy which leads to the establishment of nano oncology. Nanostructures with high specificity, are best suitable to penetrate the blood-brain barrier (BBB). Their desired physicochemical properties, such as small sizes, shape, higher surface area to volume ratio, distinctive structural features, and the possibility to attach various substances on their surface transform them into potential transport carriers able to cross various cellular and tissue barriers, including the BBB. The review emphasizes nanotechnology-based treatment strategies for the exploration of brain tumors and highlights the current progress of different nanomaterials for the effective delivery of drugs for brain tumor therapy.
Collapse
Affiliation(s)
- Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Praveen Halagali
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Amit B Patil
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Preethi Somanna
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| |
Collapse
|
16
|
Budiman A, Rusdin A, Aulifa DL. Current Techniques of Water Solubility Improvement for Antioxidant Compounds and Their Correlation with Its Activity: Molecular Pharmaceutics. Antioxidants (Basel) 2023; 12:378. [PMID: 36829937 PMCID: PMC9952677 DOI: 10.3390/antiox12020378] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/22/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
The aqueous solubility of a drug is important in the oral formulation because the drug can be absorbed from intestinal sites after being dissolved in the gastrointestinal fluid, leading to its bioavailability. Almost 80% of active pharmaceutical ingredients are poorly water-soluble, including antioxidant compounds. This makes antioxidant activity inefficient in preventing disease, particularly for orally administered formulations. Although several investigations have been carried out to improve the solubility of antioxidant compounds, there is still limited research fully discussing the subject. Therefore, this study aimed to provide an overview and discussion of the issues related to the methods that have been used to improve the solubility and activity of antioxidant compounds. Articles were found using the keywords "antioxidant" and "water solubility improvement" in the Scopus, PubMed, and Google Scholar databases. The selected articles were published within the last five years to ensure all information was up-to-date with the same objectives. The most popular methods of the strategies employed were solid dispersion, co-amorphous, and nanoparticle drug delivery systems, which were used to enhance the solubility of antioxidant compounds. These investigations produced impressive results, with a detailed discussion of the mechanism of improvement in the solubility and antioxidant activity of the compounds developed. This review shows that the strategies used to increase the solubility of antioxidant compounds successfully improved their antioxidant activity with enhanced free radical scavenging abilities.
Collapse
Affiliation(s)
- Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
| | - Agus Rusdin
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
- Department of Pharmacy, Poltekkes Kemenkes Bandung, Bandung 40161, Indonesia
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Bandung 45363, Indonesia
| |
Collapse
|
17
|
Shabani L, Abbasi M, Azarnew Z, Amani AM, Vaez A. Neuro-nanotechnology: diagnostic and therapeutic nano-based strategies in applied neuroscience. Biomed Eng Online 2023; 22:1. [PMID: 36593487 PMCID: PMC9809121 DOI: 10.1186/s12938-022-01062-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
Artificial, de-novo manufactured materials (with controlled nano-sized characteristics) have been progressively used by neuroscientists during the last several decades. The introduction of novel implantable bioelectronics interfaces that are better suited to their biological targets is one example of an innovation that has emerged as a result of advanced nanostructures and implantable bioelectronics interfaces, which has increased the potential of prostheses and neural interfaces. The unique physical-chemical properties of nanoparticles have also facilitated the development of novel imaging instruments for advanced laboratory systems, as well as intelligently manufactured scaffolds and microelectrodes and other technologies designed to increase our understanding of neural tissue processes. The incorporation of nanotechnology into physiology and cell biology enables the tailoring of molecular interactions. This involves unique interactions with neurons and glial cells in neuroscience. Technology solutions intended to effectively interact with neuronal cells, improved molecular-based diagnostic techniques, biomaterials and hybridized compounds utilized for neural regeneration, neuroprotection, and targeted delivery of medicines as well as small chemicals across the blood-brain barrier are all purposes of the present article.
Collapse
Affiliation(s)
- Leili Shabani
- grid.412571.40000 0000 8819 4698Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- grid.412571.40000 0000 8819 4698Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeynab Azarnew
- grid.412571.40000 0000 8819 4698Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- grid.412571.40000 0000 8819 4698Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- grid.412571.40000 0000 8819 4698Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
18
|
Liu HJ, Xu P. Strategies to overcome/penetrate the BBB for systemic nanoparticle delivery to the brain/brain tumor. Adv Drug Deliv Rev 2022; 191:114619. [PMID: 36372301 PMCID: PMC9724744 DOI: 10.1016/j.addr.2022.114619] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/23/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
Despite its prevalence in the management of peripheral tumors, compared to surgery and radiation therapy, chemotherapy is still a suboptimal intervention in fighting against brain cancer and cancer brain metastases. This discrepancy is mainly derived from the complicatedly physiological characteristic of intracranial tumors, including the presence of blood-brain barrier (BBB) and limited enhanced permeability and retention (EPR) effect attributed to blood-brain tumor barrier (BBTB), which largely lead to insufficient therapeutics penetrating to tumor lesions to produce pharmacological effects. Therefore, dependable methodologies that can boost the efficacy of chemotherapy for brain tumors are urgently needed. Recently, nanomedicines have shown great therapeutic potential in brain tumors by employing various transcellular strategies, paracellular strategies, and their hybrids, such as adsorptive-mediated transcytosis, receptor-mediated transcytosis, BBB disruption technology, and so on. It is compulsory to comprehensively summarize these practices to shed light on future directions in developing therapeutic regimens for brain tumors. In this review, the biological and pathological characteristics of brain tumors, including BBB and BBTB, are illustrated. After that, the emerging delivery strategies for brain tumor management are summarized into different classifications and supported with detailed examples. Finally, the potential challenges and prospects for developing and clinical application of brain tumor-oriented nanomedicine are discussed.
Collapse
Affiliation(s)
- Hai-Jun Liu
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208, USA
| | - Peisheng Xu
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208, USA.
| |
Collapse
|
19
|
Tallon C, Bell BJ, Sharma A, Pal A, Malvankar MM, Thomas AG, Yoo SW, Hollinger KR, Coleman K, Wilkinson EL, Kannan S, Haughey NJ, Kannan RM, Rais R, Slusher BS. Dendrimer-Conjugated nSMase2 Inhibitor Reduces Tau Propagation in Mice. Pharmaceutics 2022; 14:2066. [PMID: 36297501 PMCID: PMC9609094 DOI: 10.3390/pharmaceutics14102066] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/11/2022] [Accepted: 09/11/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the progressive accumulation of amyloid-β and hyperphosphorylated tau (pTau), which can spread throughout the brain via extracellular vesicles (EVs). Membrane ceramide enrichment regulated by the enzyme neutral sphingomyelinase 2 (nSMase2) is a critical component of at least one EV biogenesis pathway. Our group recently identified 2,6-Dimethoxy-4-(5-Phenyl-4-Thiophen-2-yl-1H-Imidazol-2-yl)-Phenol (DPTIP), the most potent (30 nM) and selective inhibitor of nSMase2 reported to date. However, DPTIP exhibits poor oral pharmacokinetics (PK), modest brain penetration, and rapid clearance, limiting its clinical translation. To enhance its PK properties, we conjugated DPTIP to a hydroxyl-PAMAM dendrimer delivery system, creating dendrimer-DPTIP (D-DPTIP). In an acute brain injury model, orally administered D-DPTIP significantly reduced the intra-striatal IL-1β-induced increase in plasma EVs up to 72 h post-dose, while oral DPTIP had a limited effect. In a mouse tau propagation model, where a mutant hTau (P301L/S320F) containing adeno-associated virus was unilaterally seeded into the hippocampus, oral D-DPTIP (dosed 3× weekly) significantly inhibited brain nSMase2 activity and blocked the spread of pTau to the contralateral hippocampus. These data demonstrate that dendrimer conjugation of DPTIP improves its PK properties, resulting in significant inhibition of EV propagation of pTau in mice. Dendrimer-based delivery of DPTIP has the potential to be an exciting new therapeutic for AD.
Collapse
Affiliation(s)
- Carolyn Tallon
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Benjamin J. Bell
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anjali Sharma
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Arindom Pal
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | - Seung-Wan Yoo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Kaleem Coleman
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth L. Wilkinson
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Moser Center for Leukodystrophies at Kennedy Krieger, Kennedy Krieger Institute, Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sujatha Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Hugo W. Moser Research Institute at Kennedy-Krieger Inc., Baltimore, MD 21205, USA
- Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Norman J. Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rangaramanujam M. Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Moser Center for Leukodystrophies at Kennedy Krieger, Kennedy Krieger Institute, Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
20
|
Smith GT, Radin DP, Tsirka SE. From protein-protein interactions to immune modulation: Therapeutic prospects of targeting Neuropilin-1 in high-grade glioma. Front Immunol 2022; 13:958620. [PMID: 36203599 PMCID: PMC9532003 DOI: 10.3389/fimmu.2022.958620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
In the past several years there has been a marked increase in our understanding of the pathophysiological hallmarks of glioblastoma development and progression, with specific respect to the contribution of the glioma tumor microenvironment to the rapid progression and treatment resistance of high-grade gliomas. Despite these strides, standard of care therapy still only targets rapidly dividing tumor cells in the glioma, and does little to curb the pro-tumorigenic functions of non-cancerous cells entrenched in the glioma microenvironment. This tumor promoting environment as well as the heterogeneity of high-grade gliomas contribute to the poor prognosis of this malignancy. The interaction of non-malignant cells in the microenvironment with the tumor cells accentuate phenotypes such as rapid proliferation or immunosuppression, so therapeutically modulating one target expressed on one cell type may be insufficient to restrain these rapidly developing neoplasias. With this in mind, identifying a target expressed on multiple cell types and understanding how it governs tumor-promoting functions in each cell type may have great utility in better managing this disease. Herein, we review the physiology and pathological effects of Neuropilin-1, a transmembrane co-receptor which mediates signal transduction pathways when associated with multiple other receptors. We discuss its effects on the properties of endothelial cells and on immune cell types within gliomas including glioma-associated macrophages, microglia, cytotoxic T cells and T regulatory cells. We also consider its effects when elaborated on the surface of tumor cells with respect to proliferation, stemness and treatment resistance, and review attempts to target Neuroplin-1 in the clinical setting.
Collapse
Affiliation(s)
- Gregory T. Smith
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Daniel P. Radin
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- Stony Brook Medical Scientist Training Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Stella E. Tsirka
- Molecular and Cellular Pharmacology Graduate Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- Stony Brook Medical Scientist Training Program, Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
- *Correspondence: Stella E. Tsirka,
| |
Collapse
|
21
|
Rhaman MM, Islam MR, Akash S, Mim M, Noor alam M, Nepovimova E, Valis M, Kuca K, Sharma R. Exploring the role of nanomedicines for the therapeutic approach of central nervous system dysfunction: At a glance. Front Cell Dev Biol 2022; 10:989471. [PMID: 36120565 PMCID: PMC9478743 DOI: 10.3389/fcell.2022.989471] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022] Open
Abstract
In recent decades, research scientists, molecular biologists, and pharmacologists have placed a strong emphasis on cutting-edge nanostructured materials technologies to increase medicine delivery to the central nervous system (CNS). The application of nanoscience for the treatment of neurodegenerative diseases (NDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), Huntington’s disease (HD), brain cancer, and hemorrhage has the potential to transform care. Multiple studies have indicated that nanomaterials can be used to successfully treat CNS disorders in the case of neurodegeneration. Nanomedicine development for the cure of degenerative and inflammatory diseases of the nervous system is critical. Nanoparticles may act as a drug transporter that can precisely target sick brain sub-regions, boosting therapy success. It is important to develop strategies that can penetrate the blood–brain barrier (BBB) and improve the effectiveness of medications. One of the probable tactics is the use of different nanoscale materials. These nano-based pharmaceuticals offer low toxicity, tailored delivery, high stability, and drug loading capacity. They may also increase therapeutic effectiveness. A few examples of the many different kinds and forms of nanomaterials that have been widely employed to treat neurological diseases include quantum dots, dendrimers, metallic nanoparticles, polymeric nanoparticles, carbon nanotubes, liposomes, and micelles. These unique qualities, including sensitivity, selectivity, and ability to traverse the BBB when employed in nano-sized particles, make these nanoparticles useful for imaging studies and treatment of NDs. Multifunctional nanoparticles carrying pharmacological medications serve two purposes: they improve medication distribution while also enabling cell dynamics imaging and pharmacokinetic study. However, because of the potential for wide-ranging clinical implications, safety concerns persist, limiting any potential for translation. The evidence for using nanotechnology to create drug delivery systems that could pass across the BBB and deliver therapeutic chemicals to CNS was examined in this study.
Collapse
Affiliation(s)
- Md. Mominur Rhaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mobasharah Mim
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Noor alam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Martin Valis
- Department of Neurology, Charles University in Prague, Faculty of Medicine in Hradec Králové and University Hospital, Hradec Králové, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- *Correspondence: Md. Mominur Rhaman, ; Rohit Sharma,
| |
Collapse
|
22
|
Sahoo RK, Gupta T, Batheja S, Goyal AK, Gupta U. Surface Engineered Dendrimers: A Potential Nanocarrier for the Effective Management of Glioblastoma Multiforme. Curr Drug Metab 2022; 23:708-722. [PMID: 35713127 DOI: 10.2174/1389200223666220616125524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/25/2022] [Accepted: 05/18/2022] [Indexed: 01/05/2023]
Abstract
Gliomas are the most prevailing intracranial tumors, which account for approximately 36% of the primary brain tumors of glial cells. Glioblastoma multiforme (GBM) possesses a higher degree of malignancy among different gliomas. The blood-brain barrier (BBB) protects the brain against infections and toxic substances by preventing foreign molecules or unwanted cells from entering the brain parenchyma. Nano-carriers such as liposomes, nanoparticles, dendrimers, etc. boost the brain permeability of various anticancer drugs or other drugs. The favorable properties like small size, better solubility, and the modifiable surface of dendrimers have proven their broad applicability in the better management of GBM. However, in vitro and in vivo toxicities caused by dendrimers have been a significant concern. The presence of multiple functionalities on the surface of dendrimers enables the grafting of target ligand and/or therapeutic moieties. Surface engineering improves certain properties like targeting efficiency, pharmacokinetic profile, therapeutic effect, and toxicity reduction. This review will be focused on the role of different surface-modified dendrimers in the effective management of GBM.
Collapse
Affiliation(s)
- Rakesh Kumar Sahoo
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Tanisha Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Sanya Batheja
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Amit Kumar Goyal
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| |
Collapse
|
23
|
Caraway CA, Gaitsch H, Wicks EE, Kalluri A, Kunadi N, Tyler BM. Polymeric Nanoparticles in Brain Cancer Therapy: A Review of Current Approaches. Polymers (Basel) 2022; 14:2963. [PMID: 35890738 PMCID: PMC9322801 DOI: 10.3390/polym14142963] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022] Open
Abstract
Translation of novel therapies for brain cancer into clinical practice is of the utmost importance as primary brain tumors are responsible for more than 200,000 deaths worldwide each year. While many research efforts have been aimed at improving survival rates over the years, prognosis for patients with glioblastoma and other primary brain tumors remains poor. Safely delivering chemotherapeutic drugs and other anti-cancer compounds across the blood-brain barrier and directly to tumor cells is perhaps the greatest challenge in treating brain cancer. Polymeric nanoparticles (NPs) are powerful, highly tunable carrier systems that may be able to overcome those obstacles. Several studies have shown appropriately-constructed polymeric NPs cross the blood-brain barrier, increase drug bioavailability, reduce systemic toxicity, and selectively target central nervous system cancer cells. While no studies relating to their use in treating brain cancer are in clinical trials, there is mounting preclinical evidence that polymeric NPs could be beneficial for brain tumor therapy. This review includes a variety of polymeric NPs and how their associated composition, surface modifications, and method of delivery impact their capacity to improve brain tumor therapy.
Collapse
Affiliation(s)
- Chad A. Caraway
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Hallie Gaitsch
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
- NIH-Oxford-Cambridge Scholars Program, Wellcome—MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Elizabeth E. Wicks
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
- University of Mississippi School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Anita Kalluri
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Navya Kunadi
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Betty M. Tyler
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| |
Collapse
|
24
|
Khan I, Baig MH, Mahfooz S, Imran MA, Khan MI, Dong JJ, Cho JY, Hatiboglu MA. Nanomedicine for Glioblastoma: Progress and Future Prospects. Semin Cancer Biol 2022; 86:172-186. [PMID: 35760272 DOI: 10.1016/j.semcancer.2022.06.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022]
Abstract
Glioblastoma is the most aggressive form of brain tumor, accounting for the highest mortality and morbidity rates. Current treatment for patients with glioblastoma includes maximal safe tumor resection followed by radiation therapy with concomitant temozolomide (TMZ) chemotherapy. The addition of TMZ to the conformal radiation therapy has improved the median survival time only from 12 months to 16 months in patients with glioblastoma. Despite these aggressive treatment strategies, patients' prognosis remains poor. This therapeutic failure is primarily attributed to the blood-brain barrier (BBB) that restricts the transport of TMZ from reaching the tumor site. In recent years, nanomedicine has gained considerable attention among researchers and shown promising developments in clinical applications, including the diagnosis, prognosis, and treatment of glioblastoma tumors. This review sheds light on the morphological and physiological complexity of the BBB. It also explains the development of nanomedicine strategies to enhance the permeability of drug molecules across the BBB.
Collapse
Affiliation(s)
- Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, Istanbul, Turkey
| | - Mohammad Hassan Baig
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Sadaf Mahfooz
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, Istanbul, Turkey
| | - Mohammad Azhar Imran
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Mohd Imran Khan
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Jae-June Dong
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea
| | - Jae Yong Cho
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, 120-752, Republic of Korea.
| | - Mustafa Aziz Hatiboglu
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Yalıköy St., Beykoz, Istanbul, Turkey; Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, Istanbul, Turkey.
| |
Collapse
|
25
|
Shabani L, Abbasi M, Amini M, Amani AM, Vaez A. The brilliance of nanoscience over cancer therapy: Novel promising nanotechnology-based methods for eradicating glioblastoma. J Neurol Sci 2022; 440:120316. [DOI: 10.1016/j.jns.2022.120316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 05/28/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022]
|
26
|
Naki T, Aderibigbe BA. Efficacy of Polymer-Based Nanomedicine for the Treatment of Brain Cancer. Pharmaceutics 2022; 14:1048. [PMID: 35631634 PMCID: PMC9145018 DOI: 10.3390/pharmaceutics14051048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 12/11/2022] Open
Abstract
Malignant brain tumor is a life-threatening disease with a low survival rate. The therapies available for the treatment of brain tumor is limited by poor uptake via the blood-brain barrier. The challenges with the chemotherapeutics used for the treatment of brain tumors are poor distribution, drug toxicity, and their inability to pass via the blood-brain barrier, etc. Several researchers have investigated the potential of nanomedicines for the treatment of brain cancer. Nanomedicines are designed with nanosize particle sizes with a large surface area and are loaded with bioactive agents via encapsulation, immersion, conjugation, etc. Some nanomedicines have been approved for clinical use. The most crucial part of nanomedicine is that they promote drug delivery across the blood-brain barrier, display excellent specificity, reduce drug toxicity, enhance drug bioavailability, and promote targeted drug release mechanisms. The aforementioned features make them promising therapeutics for brain targeting. This review reports the in vitro and in vivo results of nanomedicines designed for the treatment of brain cancers.
Collapse
Affiliation(s)
- Tobeka Naki
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa;
| | | |
Collapse
|
27
|
The Extension of the LeiCNS-PK3.0 Model in Combination with the "Handshake" Approach to Understand Brain Tumor Pathophysiology. Pharm Res 2022; 39:1343-1361. [PMID: 35258766 PMCID: PMC9246813 DOI: 10.1007/s11095-021-03154-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/10/2021] [Indexed: 12/22/2022]
Abstract
Micrometastatic brain tumor cells, which cause recurrence of malignant brain tumors, are often protected by the intact blood–brain barrier (BBB). Therefore, it is essential to deliver effective drugs across not only the disrupted blood-tumor barrier (BTB) but also the intact BBB to effectively treat malignant brain tumors. Our aim is to predict pharmacokinetic (PK) profiles in brain tumor regions with the disrupted BTB and the intact BBB to support the successful drug development for malignant brain tumors. LeiCNS-PK3.0, a comprehensive central nervous system (CNS) physiologically based pharmacokinetic (PBPK) model, was extended to incorporate brain tumor compartments. Most pathophysiological parameters of brain tumors were obtained from literature and two missing parameters of the BTB, paracellular pore size and expression level of active transporters, were estimated by fitting existing data, like a “handshake”. Simultaneous predictions were made for PK profiles in extracellular fluids (ECF) of brain tumors and normal-appearing brain and validated on existing data for six small molecule anticancer drugs. The LeiCNS-tumor model predicted ECF PK profiles in brain tumor as well as normal-appearing brain in rat brain tumor models and high-grade glioma patients within twofold error for most data points, in combination with estimated paracellular pore size of the BTB and active efflux clearance at the BTB. Our model demonstrated a potential to predict PK profiles of small molecule drugs in brain tumors, for which quantitative information on pathophysiological alterations is available, and contribute to the efficient and successful drug development for malignant brain tumors.
Collapse
|
28
|
Ma M, Cheng L, Wang L, Liang X, Yang L, Zhang A. Enhanced photodynamic therapy of TiO2/N-succinyl-chitosan composite for killing cancer cells. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e181116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Min Ma
- Shanxi Medical University, P. R. China
| | - Lu Cheng
- Shanxi Medical University, P. R. China
| | - Ling Wang
- Shanxi Medical University, P. R. China; Linfen Fourth People’s Hospital, P. R. China
| | | | | | | |
Collapse
|
29
|
Caro C, Avasthi A, Paez-Muñoz JM, Pernia Leal M, García-Martín ML. Passive targeting of high-grade gliomas via the EPR effect: a closed path for metallic nanoparticles? Biomater Sci 2021; 9:7984-7995. [PMID: 34710207 DOI: 10.1039/d1bm01398j] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Passive tumor targeting via the enhanced permeability and retention (EPR) effect has long been considered the most effective mechanism for the accumulation of nanoparticles inside solid tumors. However, several studies have demonstrated that the EPR effect is largely dependent on the tumor type and location. Particularly complex is the situation in brain tumors, where the presence of the blood-brain tumor barrier (BBTB) adds an extra limiting factor in reaching the tumor interstitium. However, it remains unclear whether these restraints imposed by the BBTB prevent the EPR effect from acting as an efficient tumor targeting mechanism for metallic nanoparticles. In this work, we have studied the EPR effect of metallic magnetic nanoparticles (MMNPs) in a glioblastoma (GBM) model by parametric MRI. Our results showed that only MMNPs ≤50 nm could reach the tumor interstitium, whereas larger MMNPs were unable to cross the BBTB. Furthermore, even for MMNPs around 30-50 nm, the amount of them found within the tumor was scarce and restricted to the vicinity of large tumor vessels, indicating that the BBTB strongly limits the passive accumulation of metallic nanoparticles in brain tumors. Therefore, active targeting becomes the most reasonable strategy to target metallic nanoparticles to GBMs.
Collapse
Affiliation(s)
- Carlos Caro
- BIONAND - Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía-Universidad de Málaga), C/Severo Ochoa, 35, 29590 Málaga, Spain.
| | - Ashish Avasthi
- BIONAND - Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía-Universidad de Málaga), C/Severo Ochoa, 35, 29590 Málaga, Spain.
| | - Jose M Paez-Muñoz
- BIONAND - Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía-Universidad de Málaga), C/Severo Ochoa, 35, 29590 Málaga, Spain.
| | - Manuel Pernia Leal
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain
| | - María L García-Martín
- BIONAND - Centro Andaluz de Nanomedicina y Biotecnología (Junta de Andalucía-Universidad de Málaga), C/Severo Ochoa, 35, 29590 Málaga, Spain. .,Biomedical Research Networking Center in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Spain
| |
Collapse
|
30
|
Small-Sized Co-Polymers for Targeted Delivery of Multiple Imaging and Therapeutic Agents. NANOMATERIALS 2021; 11:nano11112996. [PMID: 34835760 PMCID: PMC8625475 DOI: 10.3390/nano11112996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 12/22/2022]
Abstract
Research has increasingly focused on the delivery of high, often excessive amounts of drugs, neglecting negative aspects of the carrier's physical preconditions and biocompatibility. Among them, little attention has been paid to "small but beautiful" design of vehicle and multiple cargo to achieve effortless targeted delivery into deep tissue. The design of small biopolymers for deep tissue targeted delivery of multiple imaging agents and therapeutics (mini-nano carriers) emphasizes linear flexible polymer platforms with a hydrodynamic diameter of 4 nm to 10 nm, geometrically favoring dynamic juxtaposition of ligands to host receptors, and economic drug content. Platforms of biodegradable, non-toxic poly(β-l-malic acid) of this size carrying multiple chemically bound, optionally nature-derived or synthetic affinity peptides and drugs for a variety of purposes are described in this review with specific examples. The size, shape, and multiple attachments to membrane sites accelerate vascular escape and fast blood clearance, as well as the increase in medical treatment and contrasts for tissue imaging. High affinity antibodies routinely considered for targeting, such as the brain through the blood-brain barrier (BBB), are replaced by moderate affinity binding peptides (vectors), which penetrate at high influxes not achievable by antibodies.
Collapse
|
31
|
Kheraldine H, Rachid O, Habib AM, Al Moustafa AE, Benter IF, Akhtar S. Emerging innate biological properties of nano-drug delivery systems: A focus on PAMAM dendrimers and their clinical potential. Adv Drug Deliv Rev 2021; 178:113908. [PMID: 34390777 DOI: 10.1016/j.addr.2021.113908] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/17/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023]
Abstract
Drug delivery systems or vectors are usually needed to improve the bioavailability and effectiveness of a drug through improving its pharmacokinetics/pharmacodynamics at an organ, tissue or cellular level. However, emerging technologies with sensitive readouts as well as a greater understanding of physiological/biological systems have revealed that polymeric drug delivery systems are not biologically inert but can have innate or intrinsic biological actions. In this article, we review the emerging multiple innate biological/toxicological properties of naked polyamidoamine (PAMAM) dendrimer delivery systems in the absence of any drug cargo and discuss their correlation with the defined physicochemical properties of PAMAMs in terms of molecular size (generation), architecture, surface charge and chemistry. Further, we assess whether any of the reported intrinsic biological actions of PAMAMs such as their antimicrobial activity or their ability to sequester glucose and modulate key protein interactions or cell signaling pathways, can be exploited clinically such as in the treatment of diabetes and its complications.
Collapse
|
32
|
Doan‐Nguyen TP, Jiang S, Koynov K, Landfester K, Crespy D. Ultrasmall Nanocapsules Obtained by Controlling Ostwald Ripening. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Thao P. Doan‐Nguyen
- Max Planck-VISTEC Partner Laboratory for Sustainable Materials Vidyasirimedhi Institute of Science and Technology (VISTEC) Rayong 21210 Thailand
- Department of Materials Science and Engineering School of Molecular Science and Engineering Vidyasirimedhi Institute of Science and Technology (VISTEC) Rayong 21210 Thailand
| | - Shuai Jiang
- Max Planck-VISTEC Partner Laboratory for Sustainable Materials Vidyasirimedhi Institute of Science and Technology (VISTEC) Rayong 21210 Thailand
- Max Planck Institute for Polymer Research 55128 Mainz Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research 55128 Mainz Germany
| | | | - Daniel Crespy
- Max Planck-VISTEC Partner Laboratory for Sustainable Materials Vidyasirimedhi Institute of Science and Technology (VISTEC) Rayong 21210 Thailand
- Department of Materials Science and Engineering School of Molecular Science and Engineering Vidyasirimedhi Institute of Science and Technology (VISTEC) Rayong 21210 Thailand
| |
Collapse
|
33
|
Doan-Nguyen TP, Jiang S, Koynov K, Landfester K, Crespy D. Ultrasmall Nanocapsules Obtained by Controlling Ostwald Ripening. Angew Chem Int Ed Engl 2021; 60:18094-18102. [PMID: 34056797 DOI: 10.1002/anie.202103444] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/28/2021] [Indexed: 11/10/2022]
Abstract
We describe here a method to synthesize ultrasmall nanocapsules with a diameter of 6 nm, exhibiting a well-defined core-shell morphology. Remarkably, the nanocapules are synthesized in a miniemulsion process without the need of large amounts of surfactant as commonly used in the microemulsion process. Ultrasmall nanocapsules with an oil core and a silica shell are formed by the concurrent processes of a sol-gel reaction and Ostwald ripening. Using solvents with different water solubilities and alkoxysilanes with different reactivities, we demonstrate that sizes of obtained nanocapsules depend on the ripening rate and alkoxysilane conversion rate. The method can be also used for encapsulating natural oils such as peppermint oil and limonene. This work shows that the Ostwald ripening phenomenon can be employed beneficially for the preparation of very small colloids.
Collapse
Affiliation(s)
- Thao P Doan-Nguyen
- Max Planck-VISTEC Partner Laboratory for Sustainable Materials, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand.,Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand
| | - Shuai Jiang
- Max Planck-VISTEC Partner Laboratory for Sustainable Materials, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand.,Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | | | - Daniel Crespy
- Max Planck-VISTEC Partner Laboratory for Sustainable Materials, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand.,Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand
| |
Collapse
|
34
|
Han L, Jiang C. Evolution of blood-brain barrier in brain diseases and related systemic nanoscale brain-targeting drug delivery strategies. Acta Pharm Sin B 2021; 11:2306-2325. [PMID: 34522589 PMCID: PMC8424230 DOI: 10.1016/j.apsb.2020.11.023] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/30/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Blood–brain barrier (BBB) strictly controls matter exchange between blood and brain, and severely limits brain penetration of systemically administered drugs, resulting in ineffective drug therapy of brain diseases. However, during the onset and progression of brain diseases, BBB alterations evolve inevitably. In this review, we focus on nanoscale brain-targeting drug delivery strategies designed based on BBB evolutions and related applications in various brain diseases including Alzheimer's disease, Parkinson's disease, epilepsy, stroke, traumatic brain injury and brain tumor. The advances on optimization of small molecules for BBB crossing and non-systemic administration routes (e.g., intranasal treatment) for BBB bypassing are not included in this review.
Collapse
Key Words
- AD, Alzheimer's disease
- AMT, alpha-methyl-l-tryptophan
- Aβ, amyloid beta
- BACE1, β-secretase 1
- BBB, blood–brain barrier
- BDNF, brain derived neurotrophic factor
- BTB, blood–brain tumor barrier
- Blood–brain barrier
- Brain diseases
- Brain-targeting
- CMT, carrier-mediated transportation
- DTPA-Gd, Gd-diethyltriaminepentaacetic acid
- Drug delivery systems
- EPR, enhanced permeability and retention
- GLUT1, glucose transporter-1
- Gd, gadolinium
- ICAM-1, intercellular adhesion molecule-1
- KATP, ATP-sensitive potassium channels
- KCa, calcium-dependent potassium channels
- LAT1, L-type amino acid transporter 1
- LDL, low density lipoprotein
- LDLR, LDL receptor
- LFA-1, lymphocyte function associated antigen-1
- LRP1, LDLR-related protein 1
- MFSD2A, major facilitator superfamily domain-containing protein 2a
- MMP9, metalloproteinase-9
- MRI, magnetic resonance imaging
- NPs, nanoparticles
- Nanoparticles
- P-gp, P-glycoprotein
- PD, Parkinson's disease
- PEG, polyethyleneglycol
- PEG-PLGA, polyethyleneglycol-poly(lactic-co-glycolic acid)
- PLGA, poly(lactic-co-glycolic acid)
- PSMA, prostate-specific membrane antigen
- RAGE, receptor for advanced glycosylation end products
- RBC, red blood cell
- RMT, receptor-mediated transcytosis
- ROS, reactive oxygen species
- TBI, traumatic brain injury
- TJ, tight junction
- TfR, transferrin receptor
- VEGF, vascular endothelial growth factor
- ZO1, zona occludens 1
- siRNA, short interfering RNA
- tPA, tissue plasminogen activator
Collapse
Affiliation(s)
- Liang Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
- Corresponding author. Tel./fax: +86 512 65882089.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 200032, China
| |
Collapse
|
35
|
Wypij M, Jędrzejewski T, Trzcińska-Wencel J, Ostrowski M, Rai M, Golińska P. Green Synthesized Silver Nanoparticles: Antibacterial and Anticancer Activities, Biocompatibility, and Analyses of Surface-Attached Proteins. Front Microbiol 2021. [PMID: 33967977 DOI: 10.3389/fmicb.2021.6325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
The increasing number of multi-drug-resistant bacteria and cancer cases, that are a real threat to humankind, forces research world to develop new weapons to deal with it. Biogenic silver nanoparticles (AgNPs) are considered as a solution to this problem. Biosynthesis of AgNPs is regarded as a green, eco-friendly, low-priced process that provides small and biocompatible nanostructures with antimicrobial and anticancer activities and potential application in medicine. The biocompatibility of these nanoparticles is related to the coating with biomolecules of natural origin. The synthesis of AgNPs from actinobacterial strain was confirmed using UV-Vis spectroscopy while their morphology, crystalline structure, stability, and coating were characterized using, transmission electron microscopy (TEM), X-ray diffraction (XRD), Zeta potential and Fourier transform infrared spectroscopy (FTIR). Antibacterial activity of biogenic AgNPs was evaluated by determination of minimum inhibitory and minimum biocidal concentrations (MIC and MBC) against Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Staphylococcus aureus. The potential mechanism of antibacterial action of AgNPs was determined by measurement of ATP level. Since the use of AgNPs in biomedical applications depend on their safety, the in vitro cytotoxicity of biosynthesized AgNPs on MCF-7 human breast cancer cell line and murine macrophage cell line RAW 264.7 using MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay, cell lactate dehydrogenase (LDH) release and measurement of reactive oxygen species (ROS) level were assessed. The nanoparticle protein capping agent that can be involved in reduction of silver ions to AgNPs and their stabilization was identified using LC-MS/MS. Nanoparticles were spherical in shape, small in size (mean 13.2 nm), showed crystalline nature, good stability (-18.7 mV) and presence of capping agents. They exhibited antibacterial activity (MIC of 8-128 μg ml-1, MBC of 64-256 μg ml-1) and significantly decreased ATP levels in bacterial cells after treatment with different concentrations of AgNPs. The in vitro analysis showed that the AgNPs demonstrated dose-dependent cytotoxicity against RAW 264.7 macrophages and MCF-7 breast cancer cells but higher against the latter than the former. Cell viability decrease was found to be 42.2-14.2 and 38.0-15.5% while LDH leakage 14.6-42.7% and 19.0-45.0%, respectively. IC50 values calculated for MTT assay was found to be 16.3 and 12.0 μg ml-1 and for LDH assay 102.3 and 76.2 μg ml-1, respectively. Moreover, MCF-7 cells released a greater amount of ROS than RAW 264.7 macrophages during stimulation with all tested concentrations of AgNPs (1.47-3.13 and 1.02-2.58 fold increase, respectively). The SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) analysis revealed the presence of five protein bands at a molecular weight between 31.7 and 280.9 kDa. These proteins showed the highest homology to hypothetical proteins and porins from E. coli, Delftia sp. and Pseudomonas rhodesiae. Based on obtained results it can be concluded that biogenic AgNPs were capped with proteins and demonstrated potential as antimicrobial and anticancer agent.
Collapse
Affiliation(s)
- Magdalena Wypij
- Department of Microbiology, Nicolaus Copernicus University, Toruń, Poland
| | | | | | - Maciej Ostrowski
- Department of Biochemistry, Nicolaus Copernicus University, Toruń, Poland
| | - Mahendra Rai
- Department of Microbiology, Nicolaus Copernicus University, Toruń, Poland.,Nanobiotechnology Laboratory, Department of Biotechnology, SGB Amravati University, Amravati, India
| | - Patrycja Golińska
- Department of Microbiology, Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
36
|
Zhang W, Mehta A, Tong Z, Esser L, Voelcker NH. Development of Polymeric Nanoparticles for Blood-Brain Barrier Transfer-Strategies and Challenges. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003937. [PMID: 34026447 PMCID: PMC8132167 DOI: 10.1002/advs.202003937] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/20/2020] [Indexed: 05/04/2023]
Abstract
Neurological disorders such as Alzheimer's disease, stroke, and brain cancers are difficult to treat with current drugs as their delivery efficacy to the brain is severely hampered by the presence of the blood-brain barrier (BBB). Drug delivery systems have been extensively explored in recent decades aiming to circumvent this barrier. In particular, polymeric nanoparticles have shown enormous potentials owing to their unique properties, such as high tunability, ease of synthesis, and control over drug release profile. However, careful analysis of their performance in effective drug transport across the BBB should be performed using clinically relevant testing models. In this review, polymeric nanoparticle systems for drug delivery to the central nervous system are discussed with an emphasis on the effects of particle size, shape, and surface modifications on BBB penetration. Moreover, the authors critically analyze the current in vitro and in vivo models used to evaluate BBB penetration efficacy, including the latest developments in the BBB-on-a-chip models. Finally, the challenges and future perspectives for the development of polymeric nanoparticles to combat neurological disorders are discussed.
Collapse
Affiliation(s)
- Weisen Zhang
- Drug Delivery, Disposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
| | - Ami Mehta
- Drug Delivery, Disposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- IITB Monash Research AcademyBombayMumbai400076India
| | - Ziqiu Tong
- Drug Delivery, Disposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
| | - Lars Esser
- Drug Delivery, Disposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVIC3168Australia
| | - Nicolas H. Voelcker
- Drug Delivery, Disposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVIC3168Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication FacilityClaytonVIC3168Australia
- Department of Materials Science and EngineeringMonash UniversityClaytonVIC3800Australia
| |
Collapse
|
37
|
Wypij M, Jędrzejewski T, Trzcińska-Wencel J, Ostrowski M, Rai M, Golińska P. Green Synthesized Silver Nanoparticles: Antibacterial and Anticancer Activities, Biocompatibility, and Analyses of Surface-Attached Proteins. Front Microbiol 2021; 12:632505. [PMID: 33967977 PMCID: PMC8100210 DOI: 10.3389/fmicb.2021.632505] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
The increasing number of multi-drug-resistant bacteria and cancer cases, that are a real threat to humankind, forces research world to develop new weapons to deal with it. Biogenic silver nanoparticles (AgNPs) are considered as a solution to this problem. Biosynthesis of AgNPs is regarded as a green, eco-friendly, low-priced process that provides small and biocompatible nanostructures with antimicrobial and anticancer activities and potential application in medicine. The biocompatibility of these nanoparticles is related to the coating with biomolecules of natural origin. The synthesis of AgNPs from actinobacterial strain was confirmed using UV-Vis spectroscopy while their morphology, crystalline structure, stability, and coating were characterized using, transmission electron microscopy (TEM), X-ray diffraction (XRD), Zeta potential and Fourier transform infrared spectroscopy (FTIR). Antibacterial activity of biogenic AgNPs was evaluated by determination of minimum inhibitory and minimum biocidal concentrations (MIC and MBC) against Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, and Staphylococcus aureus. The potential mechanism of antibacterial action of AgNPs was determined by measurement of ATP level. Since the use of AgNPs in biomedical applications depend on their safety, the in vitro cytotoxicity of biosynthesized AgNPs on MCF-7 human breast cancer cell line and murine macrophage cell line RAW 264.7 using MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay, cell lactate dehydrogenase (LDH) release and measurement of reactive oxygen species (ROS) level were assessed. The nanoparticle protein capping agent that can be involved in reduction of silver ions to AgNPs and their stabilization was identified using LC-MS/MS. Nanoparticles were spherical in shape, small in size (mean 13.2 nm), showed crystalline nature, good stability (-18.7 mV) and presence of capping agents. They exhibited antibacterial activity (MIC of 8-128 μg ml-1, MBC of 64-256 μg ml-1) and significantly decreased ATP levels in bacterial cells after treatment with different concentrations of AgNPs. The in vitro analysis showed that the AgNPs demonstrated dose-dependent cytotoxicity against RAW 264.7 macrophages and MCF-7 breast cancer cells but higher against the latter than the former. Cell viability decrease was found to be 42.2-14.2 and 38.0-15.5% while LDH leakage 14.6-42.7% and 19.0-45.0%, respectively. IC50 values calculated for MTT assay was found to be 16.3 and 12.0 μg ml-1 and for LDH assay 102.3 and 76.2 μg ml-1, respectively. Moreover, MCF-7 cells released a greater amount of ROS than RAW 264.7 macrophages during stimulation with all tested concentrations of AgNPs (1.47-3.13 and 1.02-2.58 fold increase, respectively). The SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) analysis revealed the presence of five protein bands at a molecular weight between 31.7 and 280.9 kDa. These proteins showed the highest homology to hypothetical proteins and porins from E. coli, Delftia sp. and Pseudomonas rhodesiae. Based on obtained results it can be concluded that biogenic AgNPs were capped with proteins and demonstrated potential as antimicrobial and anticancer agent.
Collapse
Affiliation(s)
- Magdalena Wypij
- Department of Microbiology, Nicolaus Copernicus University, Toruń, Poland
| | | | | | - Maciej Ostrowski
- Department of Biochemistry, Nicolaus Copernicus University, Toruń, Poland
| | - Mahendra Rai
- Department of Microbiology, Nicolaus Copernicus University, Toruń, Poland
- Nanobiotechnology Laboratory, Department of Biotechnology, SGB Amravati University, Amravati, India
| | - Patrycja Golińska
- Department of Microbiology, Nicolaus Copernicus University, Toruń, Poland
| |
Collapse
|
38
|
Balakrishnan PB, Sweeney EE. Nanoparticles for Enhanced Adoptive T Cell Therapies and Future Perspectives for CNS Tumors. Front Immunol 2021; 12:600659. [PMID: 33833751 PMCID: PMC8021848 DOI: 10.3389/fimmu.2021.600659] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/08/2021] [Indexed: 12/20/2022] Open
Abstract
Adoptive T cell therapy has emerged as a revolutionary immunotherapy for treating cancer. Despite immense promise and clinical success in some hematologic malignancies, limitations remain that thwart its efficacy in solid tumors. Particularly in tumors of the central nervous system (CNS), T cell therapy is often restricted by the difficulty in intratumoral delivery across anatomical niches, suboptimal T cell specificity or activation, and intratumoral T cell dysfunction due to immunosuppressive tumor microenvironments (TMEs). Nanoparticles may offer several advantages to overcome these limitations of T cell therapy, as they can be designed to robustly and specifically activate T cells ex vivo prior to adoptive transfer, to encapsulate T cell stimulating agents for co-localized stimulation, and to be conjugated onto T cells for added functionality. This perspective highlights recent preclinical advances in using nanoparticles to enhance T cell therapy, and discusses the potential applicability and constraints of nanoparticle-enhanced T cells as a new platform for treating CNS tumors.
Collapse
Affiliation(s)
- Preethi Bala Balakrishnan
- The George Washington University Cancer Center, School of Medicine and Health Sciences, George Washington University, Washington, DC, United States
| | - Elizabeth E Sweeney
- The George Washington University Cancer Center, School of Medicine and Health Sciences, George Washington University, Washington, DC, United States
| |
Collapse
|
39
|
Perrelli A, Fatehbasharzad P, Benedetti V, Ferraris C, Fontanella M, De Luca E, Moglianetti M, Battaglia L, Retta SF. Towards precision nanomedicine for cerebrovascular diseases with emphasis on Cerebral Cavernous Malformation (CCM). Expert Opin Drug Deliv 2021; 18:849-876. [PMID: 33406376 DOI: 10.1080/17425247.2021.1873273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Cerebrovascular diseases encompass various disorders of the brain vasculature, such as ischemic/hemorrhagic strokes, aneurysms, and vascular malformations, also affecting the central nervous system leading to a large variety of transient or permanent neurological disorders. They represent major causes of mortality and long-term disability worldwide, and some of them can be inherited, including Cerebral Cavernous Malformation (CCM), an autosomal dominant cerebrovascular disease linked to mutations in CCM1/KRIT1, CCM2, or CCM3/PDCD10 genes.Areas covered: Besides marked clinical and etiological heterogeneity, some commonalities are emerging among distinct cerebrovascular diseases, including key pathogenetic roles of oxidative stress and inflammation, which are increasingly recognized as major disease hallmarks and therapeutic targets. This review provides a comprehensive overview of the different clinical features and common pathogenetic determinants of cerebrovascular diseases, highlighting major challenges, including the pressing need for new diagnostic and therapeutic strategies, and focusing on emerging innovative features and promising benefits of nanomedicine strategies for early detection and targeted treatment of such diseases.Expert opinion: Specifically, we describe and discuss the multiple physico-chemical features and unique biological advantages of nanosystems, including nanodiagnostics, nanotherapeutics, and nanotheranostics, that may help improving diagnosis and treatment of cerebrovascular diseases and neurological comorbidities, with an emphasis on CCM disease.
Collapse
Affiliation(s)
- Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| | - Parisa Fatehbasharzad
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| | - Valerio Benedetti
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| | - Chiara Ferraris
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, Torino, Italy
| | - Marco Fontanella
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Elisa De Luca
- Nanobiointeractions & Nanodiagnostics, Center for Biomolecular Nanotechnologies, Arnesano, Lecce, Italy.,Institute for Microelectronics and Microsystems (IMM), CNR, Lecce, Italy
| | - Mauro Moglianetti
- Nanobiointeractions & Nanodiagnostics, Center for Biomolecular Nanotechnologies, Arnesano, Lecce, Italy.,Istituto Italiano Di Tecnologia, Nanobiointeractions & Nanodiagnostics, Genova, Italy
| | - Luigi Battaglia
- Department of Drug Science and Technology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, University of Torino, Torino, Italy
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy.,CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Orbassano, Torino Italy
| |
Collapse
|
40
|
Saleh MAA, de Lange ECM. Impact of CNS Diseases on Drug Delivery to Brain Extracellular and Intracellular Target Sites in Human: A "WHAT-IF" Simulation Study. Pharmaceutics 2021; 13:95. [PMID: 33451111 PMCID: PMC7828633 DOI: 10.3390/pharmaceutics13010095] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/23/2022] Open
Abstract
The blood-brain barrier (BBB) is equipped with unique physical and functional processes that control central nervous system (CNS) drug transport and the resulting concentration-time profiles (PK). In CNS diseases, the altered BBB and CNS pathophysiology may affect the CNS PK at the drug target sites in the brain extracellular fluid (brainECF) and intracellular fluid (brainICF) that may result in changes in CNS drug effects. Here, we used our human CNS physiologically-based PK model (LeiCNS-PK3.0) to investigate the impact of altered cerebral blood flow (CBF), tight junction paracellular pore radius (pararadius), brainECF volume, and pH of brainECF (pHECF) and of brainICF (pHICF) on brainECF and brainICF PK for 46 small drugs with distinct physicochemical properties. LeiCNS-PK3.0 simulations showed a drug-dependent effect of the pathophysiological changes on the rate and extent of BBB transport and on brainECF and brainICF PK. Altered pararadius, pHECF, and pHICF affected both the rate and extent of BBB drug transport, whereas changes in CBF and brainECF volume modestly affected the rate of BBB drug transport. While the focus is often on BBB paracellular and active transport processes, this study indicates that also changes in pH should be considered for their important implications on brainECF and brainICF target site PK.
Collapse
Affiliation(s)
| | - Elizabeth C. M. de Lange
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands;
| |
Collapse
|
41
|
Saw PE, Xu X, Kang BR, Lee J, Lee YS, Kim C, Kim H, Kang SH, Na YJ, Moon HJ, Kim JH, Park YK, Yoon W, Kim JH, Kwon TH, Choi C, Jon S, Chong K. Extra-domain B of fibronectin as an alternative target for drug delivery and a cancer diagnostic and prognostic biomarker for malignant glioma. Am J Cancer Res 2021; 11:941-957. [PMID: 33391514 PMCID: PMC7738868 DOI: 10.7150/thno.44948] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022] Open
Abstract
Extra-domain B of fibronectin (EDB-FN) is an alternatively spliced form of fibronectin with high expression in the extracellular matrix of neovascularized tissues and malignant cancer cells. In this study, we evaluated the practicality of using EDB-FN as a biomarker and therapeutic target for malignant gliomas (MGs), representative intractable diseases involving brain tumors. Methods: The microarray- and sequence-based patient transcriptomic database 'Oncopression' and tissue microarray of MG patient tissue samples were analyzed. EDB-FN data were extracted and evaluated from 23,344 patient samples of 17 types of cancer to assess its effectiveness and selectivity as a molecular target. To strengthen the results of the patient data analysis, the utility of EDB-FN as a molecular marker and target for MG was verified using active EDB-FN-targeting ultrasmall lipidic micellar nanoparticles (~12 nm), which had a high drug-loading capacity and were efficiently internalized by MG cells in vitro and in vivo. Results: Brain tumors had a 1.42-fold cancer-to-normal ratio (p < 0.0001), the second highest among 17 cancers after head and neck cancer. Patient tissue microarray analysis showed that the EDB-FN high-expression group had a 5.5-fold higher risk of progression than the EDB-FN low-expression group (p < 0.03). By labeling docetaxel-containing ultrasmall micelles with a bipodal aptide targeting EDB-FN (termed APTEDB-DSPE-DTX), we generated micelles that could specifically bind to MG cells, leading to superior antitumor efficacy of EDB-FN-targeting nanoparticles compared to nontargeting controls. Conclusions: Taken together, these results show that EDB-FN can be an effective drug delivery target and biomarker for MG.
Collapse
|
42
|
Giuliano K, Etchill E, Zhou X, Lui C, Suarez-Pierre A, Sharma R, Wilson MA, Blue ME, Troncoso JC, Kannan S, Johnston MV, Sharma A, Kannan RM, Baumgartner WA, Lawton J. NMDA Receptor Antagonism for Neuroprotection in a Canine Model of Hypothermic Circulatory Arrest. J Surg Res 2020; 260:177-189. [PMID: 33348169 DOI: 10.1016/j.jss.2020.11.075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Hypothermic circulatory arrest (HCA) is associated with neurologic morbidity, in part mediated by activation of the N-methyl-D-aspartate glutamate receptor causing excitotoxicity and neuronal apoptosis. Using a canine model, we hypothesized that the N-methyl-D-aspartate receptor antagonist MK801 would provide neuroprotection and that MK801 conjugation to dendrimer nanoparticles would improve efficacy. MATERIALS AND METHODS Male hound dogs were placed on cardiopulmonary bypass, cooled to 18°C, and underwent 90 min of HCA. Dendrimer conjugates (d-MK801) were prepared by covalently linking dendrimer surface OH groups to MK801. Six experimental groups received either saline (control), medium- (0.15 mg/kg) or high-dose (1.56 mg/kg) MK801, or low- (0.05 mg/kg), medium-, or high-dose d-MK801. At 24, 48, and 72 h after HCA, animals were scored by a standardized neurobehavioral paradigm (higher scores indicate increasing deficits). Cerebrospinal fluid was obtained at baseline, eight, 24, 48, and 72 h after HCA. At 72 h, brains were examined for histopathologic injury in a blinded manner (higher scores indicate more injury). RESULTS Neurobehavioral deficit scores were reduced by low-dose d-MK801 on postoperative day two (P < 0.05) and by medium-dose d-MK801 on postoperative day 3 (P = 0.05) compared with saline controls, but free drug had no effect. In contrast, high-dose free MK801 significantly improved histopathology scores compared with saline (P < 0.05) and altered biomarkers of injury in cerebrospinal fluid, with a significant reduction in phosphorylated neurofilament-H for high-dose MK801 versus saline (P < 0.05). CONCLUSIONS Treatment with MK-801 demonstrated significant improvement in neurobehavioral and histopathology scores after HCA, although not consistently across doses and conjugates.
Collapse
Affiliation(s)
- Katherine Giuliano
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eric Etchill
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xun Zhou
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cecillia Lui
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alejandro Suarez-Pierre
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rishi Sharma
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mary Ann Wilson
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland
| | - Mary E Blue
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland
| | - Juan C Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sujatha Kannan
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael V Johnston
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland
| | - Anjali Sharma
- Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Willian A Baumgartner
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jennifer Lawton
- Division of Cardiac Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
43
|
Blood-Brain Barrier Modulation to Improve Glioma Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12111085. [PMID: 33198244 PMCID: PMC7697580 DOI: 10.3390/pharmaceutics12111085] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
The blood-brain barrier (BBB) is formed by brain microvascular endothelial cells that are sealed by tight junctions, making it a significant obstacle for most brain therapeutics. The poor BBB penetration of newly developed therapeutics has therefore played a major role in limiting their clinical success. A particularly challenging therapeutic target is glioma, which is the most frequently occurring malignant brain tumor. Thus, to enhance therapeutic uptake in tumors, researchers have been developing strategies to modulate BBB permeability. However, most conventional BBB opening strategies are difficult to apply in the clinical setting due to their broad, non-specific modulation of the BBB, which can result in damage to normal brain tissue. In this review, we have summarized strategies that could potentially be used to selectively and efficiently modulate the tumor BBB for more effective glioma treatment.
Collapse
|
44
|
Ortiz N, Vásquez PA, Vidal F, Díaz CF, Guzmán JL, Jiménez VA, Alderete JB. Polyamidoamine-based nanovector for the efficient delivery of methotrexate to U87 glioma cells. Nanomedicine (Lond) 2020; 15:2771-2784. [PMID: 33073670 DOI: 10.2217/nnm-2020-0305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The purpose of this study was to design a polyamidoamine (PAMAM)-based nanovector for the efficient delivery of methotrexate to U87 glioma cells. To this end, 0-100% acetylated PAMAM dendrimers of the fourth generation were synthesized and evaluated using drug encapsulation measurements, molecular dynamics simulations, neurotoxicity assays and neuronal internalization experiments. The best system was tested as a nanovector for methotrexate delivery to U87 glioma cells. The authors found that 25% acetylated PAMAM dendrimers of the fourth-generation combine low intrinsic toxicity, large drug complexation capacity and efficient internalization into hippocampal neurons. Nanovector complexation enhances the cytotoxic response of methotrexate against U87 glioma cells compared with free drug solutions. In conclusion, 25% acetylated PAMAM dendrimers of the fourth-generation increase drug uptake by glioma cells and thereby act as efficient nanovectors for methotrexate delivery.
Collapse
Affiliation(s)
- Natalia Ortiz
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile
| | - Pilar A Vásquez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile
| | - Felipe Vidal
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile
| | - Carola F Díaz
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Talcahuano 4260000, Chile
| | - José L Guzmán
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4030000, Chile
| | - Verónica A Jiménez
- Departamento de Ciencias Químicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Talcahuano 4260000, Chile
| | - Joel B Alderete
- Instituto de Química de Recursos Naturales, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
45
|
Abstract
Background:
Drug delivery to cancerous brain is a challenging task as it is
surrounded by an efficient protective barrier. The main hurdles for delivery of bioactive
molecules to cancerous brain are blood brain barrier (BBB), the invasive nature of gliomas,
drug resistance, and difficult brain interstitium transportation. Therefore, treatment
of brain cancer with the available drug regimen is difficult and has shown little improvement
in recent years.
Methods:
We searched about recent advancements in the use of nanomedicine for effective
treatment of the brain cancer. We focused on the use of liposomes, nanoparticles,
polymeric micelles, and dendrimers to improve brain cancer therapy.
Results:
Nanomedicines are well suited for the treatment of brain cancer owing to their
highly acceptable biological, chemical, and physical properties. Smaller size of nanomedicines
also enhances their anticancer potential and penetration into blood brain barrier
(BBB).
Conclusion:
Recently, nanomedicine based approaches have been developed and investigated
for effective treatment of brain cancer. Some of these have been translated into
clinical practice, in order to attain therapeutic needs of gliomas. Future advancements in
nanomedicines will likely produce significant changes in methods and practice of brain
cancer therapy.
Collapse
Affiliation(s)
- Shivani Verma
- I. K. Gujral Punjab Technical University, Jalandhar-Punjab 144601, India
| | - Puneet Utreja
- I. K. Gujral Punjab Technical University, Jalandhar-Punjab 144601, India
| | - Lalit Kumar
- I. K. Gujral Punjab Technical University, Jalandhar-Punjab 144601, India
| |
Collapse
|
46
|
Wypij M, Jędrzejewski T, Ostrowski M, Trzcińska J, Rai M, Golińska P. Biogenic Silver Nanoparticles: Assessment of Their Cytotoxicity, Genotoxicity and Study of Capping Proteins. Molecules 2020; 25:E3022. [PMID: 32630696 PMCID: PMC7412363 DOI: 10.3390/molecules25133022] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/02/2023] Open
Abstract
The development of nanotechnology in the last two decades has led to the use of silver nanoparticles (AgNPs) in various biomedical applications, including antimicrobial, anti-inflammatory, and anticancer therapies. However, the potential of the medical application of AgNPs depends on the safety of their use. In this work, we assessed the in vitro cytotoxicity and genotoxicity of silver nanoparticles and identified biomolecules covering AgNPs synthesized from actinobacterial strain SH11. The cytotoxicity of AgNPs against MCF-7 human breast cancer cell line and murine macrophage cell line RAW 264.7 was studied by MTT assay, cell LDH (lactate dehydrogenase) release, and the measurement of ROS (reactive oxygen species) level while genotoxicity in Salmonella typhimurium cells was testing using the Ames test. The in vitro analysis showed that the tested nanoparticles demonstrated dose-dependent cytotoxicity against RAW 264.6 macrophages and MCF-7 breast cancer cells. Moreover, biosynthesized AgNPs did not show a mutagenic effect of S. typhimurium. The analyses and identification of biomolecules present on the surface of silver nanoparticles showed that they were associated with proteins. The SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) analysis revealed the presence of 34 and 43 kDa protein bands. The identification of proteins performed by using LC-MS/MS (liquid chromatography with tandem mass spectrometry) demonstrated their highest homology to bacterial porins. Capping biomolecules of natural origin may be involved in the synthesis process of AgNPs or may be responsible for their stabilization. Moreover, the presence of natural proteins on the surface of bionanoparticles eliminates the postproduction steps of capping which is necessary for chemical synthesis to obtain the stable nanostructures required for application in medicine.
Collapse
Affiliation(s)
- Magdalena Wypij
- Department of Microbiology, Nicolaus Copernicus University, Lwowska 1, 87100 Torun, Poland;
| | - Tomasz Jędrzejewski
- Department of Immunology, Nicolaus Copernicus University, Lwowska 1, 87100 Torun, Poland;
| | - Maciej Ostrowski
- Department of Biochemistry, Nicolaus Copernicus University, Lwowska 1, 87100 Torun, Poland;
| | - Joanna Trzcińska
- Department of Microbiology, Nicolaus Copernicus University, Lwowska 1, 87100 Torun, Poland;
| | - Mahendra Rai
- Nanobiotechnology Lab., Department of Biotechnology, SGB Amravati University, Amravati, Maharashtra 444602, India;
| | - Patrycja Golińska
- Department of Microbiology, Nicolaus Copernicus University, Lwowska 1, 87100 Torun, Poland;
| |
Collapse
|
47
|
Fadzen CM, Wolfe JM, Zhou W, Cho CF, von Spreckelsen N, Hutchinson KT, Lee YC, Chiocca EA, Lawler SE, Yilmaz OH, Lippard SJ, Pentelute BL. A Platinum(IV) Prodrug-Perfluoroaryl Macrocyclic Peptide Conjugate Enhances Platinum Uptake in the Brain. J Med Chem 2020; 63:6741-6747. [PMID: 32410451 DOI: 10.1021/acs.jmedchem.0c00022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Effective delivery to the brain limits the development of novel glioblastoma therapies. Here, we introduce conjugation between platinum(IV) prodrugs of cisplatin and perfluoroaryl peptide macrocycles to increase brain uptake. We demonstrate that one such conjugate shows efficacy against glioma stem-like cells. We investigate the pharmacokinetics of this conjugate in mice and show that the amount of platinum in the brain after treatment with the conjugate is 15-fold greater than with cisplatin after 5 h.
Collapse
Affiliation(s)
- Colin M Fadzen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Justin M Wolfe
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Wen Zhou
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Choi-Fong Cho
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Niklas von Spreckelsen
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States.,Department of Neurosurgery, Center for Neurosurgery, Faculty of Medicine and University Hospital, University of Cologne 50937 Cologne, Germany
| | - Kathryn T Hutchinson
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yen-Chun Lee
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - E Antonio Chiocca
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sean E Lawler
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Omer H Yilmaz
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Stephen J Lippard
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bradley L Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States.,The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
48
|
Trigonelline-loaded chitosan nanoparticles prompted antitumor activity on glioma cells and biocompatibility with pheochromocytoma cells. Int J Biol Macromol 2020; 163:36-43. [PMID: 32585274 DOI: 10.1016/j.ijbiomac.2020.06.165] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 11/21/2022]
Abstract
Trigonelline-loaded water-soluble chitosan nanoparticles (Trigo-WSCS NPs) were prepared for the treatment of glioblastoma (targeting C6 glioma cells) and also evaluated its biocompatibility with rat adrenal pheochromocytoma cells (PC12 cells). WSCS-Trigo NPs characteristics were determined using UV-Visible spectrophotometer, FTIR, XRD, TEM, DLS, and Zeta potential. Trigo-WSCS NPs were noted to have a spherical shape, with an average size of 356 nm. Trigo-WSCS NPs zeta potential was 30.9 mv, which expresses its good stability. The WSCS-Trigo NPs considerably inhibited the growth of rat C6 glioma cells and exhibited an IC50 concentration of 34 μg/mL. Further, Trigo-WSCS NPs were biocompatible with PC12 cells in terms of enhancing neurite growth and differentiation. In conclusion, Trigo-WSCS NPs could act as an antitumor drug for the treatment of glioblastoma as suggested by the in vitro studies.
Collapse
|
49
|
Belykh E, Shaffer KV, Lin C, Byvaltsev VA, Preul MC, Chen L. Blood-Brain Barrier, Blood-Brain Tumor Barrier, and Fluorescence-Guided Neurosurgical Oncology: Delivering Optical Labels to Brain Tumors. Front Oncol 2020; 10:739. [PMID: 32582530 PMCID: PMC7290051 DOI: 10.3389/fonc.2020.00739] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/17/2020] [Indexed: 12/17/2022] Open
Abstract
Recent advances in maximum safe glioma resection have included the introduction of a host of visualization techniques to complement intraoperative white-light imaging of tumors. However, barriers to the effective use of these techniques within the central nervous system remain. In the healthy brain, the blood-brain barrier ensures the stability of the sensitive internal environment of the brain by protecting the active functions of the central nervous system and preventing the invasion of microorganisms and toxins. Brain tumors, however, often cause degradation and dysfunction of this barrier, resulting in a heterogeneous increase in vascular permeability throughout the tumor mass and outside it. Thus, the characteristics of both the blood-brain and blood-brain tumor barriers hinder the vascular delivery of a variety of therapeutic substances to brain tumors. Recent developments in fluorescent visualization of brain tumors offer improvements in the extent of maximal safe resection, but many of these fluorescent agents must reach the tumor via the vasculature. As a result, these fluorescence-guided resection techniques are often limited by the extent of vascular permeability in tumor regions and by the failure to stain the full volume of tumor tissue. In this review, we describe the structure and function of both the blood-brain and blood-brain tumor barriers in the context of the current state of fluorescence-guided imaging of brain tumors. We discuss features of currently used techniques for fluorescence-guided brain tumor resection, with an emphasis on their interactions with the blood-brain and blood-tumor barriers. Finally, we discuss a selection of novel preclinical techniques that have the potential to enhance the delivery of therapeutics to brain tumors in spite of the barrier properties of the brain.
Collapse
Affiliation(s)
- Evgenii Belykh
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Kurt V. Shaffer
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Chaoqun Lin
- Department of Neurosurgery, School of Medicine, Southeast University, Nanjing, China
| | - Vadim A. Byvaltsev
- Department of Neurosurgery, Irkutsk State Medical University, Irkutsk, Russia
| | - Mark C. Preul
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
50
|
Houston Z, Bunt J, Chen KS, Puttick S, Howard CB, Fletcher NL, Fuchs AV, Cui J, Ju Y, Cowin G, Song X, Boyd AW, Mahler SM, Richards LJ, Caruso F, Thurecht KJ. Understanding the Uptake of Nanomedicines at Different Stages of Brain Cancer Using a Modular Nanocarrier Platform and Precision Bispecific Antibodies. ACS CENTRAL SCIENCE 2020; 6:727-738. [PMID: 32490189 PMCID: PMC7256936 DOI: 10.1021/acscentsci.9b01299] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Indexed: 06/11/2023]
Abstract
Increasing accumulation and retention of nanomedicines within tumor tissue is a significant challenge, particularly in the case of brain tumors where access to the tumor through the vasculature is restricted by the blood-brain barrier (BBB). This makes the application of nanomedicines in neuro-oncology often considered unfeasible, with efficacy limited to regions of significant disease progression and compromised BBB. However, little is understood about how the evolving tumor-brain physiology during disease progression affects the permeability and retention of designer nanomedicines. We report here the development of a modular nanomedicine platform that, when used in conjunction with a unique model of how tumorigenesis affects BBB integrity, allows investigation of how nanomaterial properties affect uptake and retention in brain tissue. By combining different in vivo longitudinal imaging techniques (including positron emission tomography and magnetic resonance imaging), we have evaluated the retention of nanomedicines with predefined physicochemical properties (size and surface functionality) and established a relationship between structure and tissue accumulation as a function of a new parameter that measures BBB leakiness; this offers significant advancements in our ability to relate tumor accumulation of nanomedicines to more physiologically relevant parameters. Our data show that accumulation of nanomedicines in brain tumor tissue is better correlated with the leakiness of the BBB than actual tumor volume. This was evaluated by establishing brain tumors using a spontaneous and endogenously derived glioblastoma model providing a unique opportunity to assess these parameters individually and compare the results across multiple mice. We also quantitatively demonstrate that smaller nanomedicines (20 nm) can indeed cross the BBB and accumulate in tumors at earlier stages of the disease than larger analogues, therefore opening the possibility of developing patient-specific nanoparticle treatment interventions in earlier stages of the disease. Importantly, these results provide a more predictive approach for designing efficacious personalized nanomedicines based on a particular patient's condition.
Collapse
Affiliation(s)
- Zachary
H. Houston
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jens Bunt
- Queensland
Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kok-Siong Chen
- Queensland
Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
- Brigham
and Women’s Hospital, Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Simon Puttick
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- Commonwealth
Scientific and Industrial Research Organisation, Probing Biosystems
Future Science Platform, Royal Brisbane
and Women’s Hospital, Brisbane, Queensland 4029, Australia
| | - Christopher B. Howard
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training
Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training Centre for Biopharmaceutical
Innovation The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas L. Fletcher
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Adrian V. Fuchs
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jiwei Cui
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- Key
Laboratory of Colloid and Interface Chemistry of the Ministry of Education,
School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, China
| | - Yi Ju
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Gary Cowin
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
| | - Xin Song
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
| | - Andrew W. Boyd
- Leukaemia
Foundation Laboratory, QIMR-Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
- Department
of Medicine, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Stephen M. Mahler
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training Centre for Biopharmaceutical
Innovation The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Linda J. Richards
- Queensland
Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
- The
School of Biomedical Sciences, The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Frank Caruso
- Department
of Chemical Engineering, The University
of Melbourne, Parkville, Victoria 3010, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kristofer J. Thurecht
- Centre
for Advanced Imaging, The University of
Queensland, St Lucia, Queensland 4072, Australia
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC
Centre of Excellence in Convergent BioNano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
- ARC Training
Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|