1
|
Cho S, Jo H, Hwang YJ, Kim C, Jo YH, Yun JW. Potential impact of underlying diseases influencing ADME in nonclinical safety assessment. Food Chem Toxicol 2024; 188:114636. [PMID: 38582343 DOI: 10.1016/j.fct.2024.114636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
Nonclinical studies involve in vitro, in silico, and in vivo experiments to assess the toxicokinetics, toxicology, and safety pharmacology of drugs according to regulatory requirements by a national or international authority. In this review, we summarize the potential effects of various underlying diseases governing the absorption, distribution, metabolism, and excretion (ADME) of drugs to consider the use of animal models of diseases in nonclinical trials. Obesity models showed alterations in hepatic metabolizing enzymes, transporters, and renal pathophysiology, which increase the risk of drug-induced toxicity. Diabetes models displayed changes in hepatic metabolizing enzymes, transporters, and glomerular filtration rates (GFR), leading to variability in drug responses and susceptibility to toxicity. Animal models of advanced age exhibited impairment of drug metabolism and kidney function, thereby reducing the drug-metabolizing capacity and clearance. Along with changes in hepatic metabolic enzymes, animal models of metabolic syndrome-related hypertension showed renal dysfunction, resulting in a reduced GFR and urinary excretion of drugs. Taken together, underlying diseases can induce dysfunction of organs involved in the ADME of drugs, ultimately affecting toxicity. Therefore, the use of animal models of representative underlying diseases in nonclinical toxicity studies can be considered to improve the predictability of drug side effects before clinical trials.
Collapse
Affiliation(s)
- Sumin Cho
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Harin Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeon Jeong Hwang
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Changuk Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Yong Hyeon Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
2
|
Bak A, Kozik V, Swietlicka A, Baran W, Smolinski A, Zięba A. Towards Symmetric Thioamides: Microwave-Aided Synthesis of Terephthalic Acid Derivatives. Pharmaceuticals (Basel) 2023; 16:984. [PMID: 37513896 PMCID: PMC10385826 DOI: 10.3390/ph16070984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The multistep synthesis of novel bis-terephthalthioamides based on methyl esters of amino acids (AAs) was proposed using conventional heating and microwave-assisted approaches. In fact, the comparative case study on the thionation of new symmetrical diamides with Lawesson's reagent (LR) was performed. The microwave-accelerated small-scale methodology was successfully employed on the whole pathway from substrates (Gly, Ala, Val, Tyr, Ser) to products (symmetrical dithioamides of terephthalic acid), resulting in significantly reduced reaction time, energy requirements, and slightly increased reaction yields when compared to conventional heating. Moreover, the intermolecular similarity of novel terephthalic acid derivatives was estimated in the multidimensional space (mDS) of the structure/property-related in silico descriptors using principal component analysis (PCA) and hierarchical clustering analysis (HCA). The distance-oriented structure/property distribution was also correlated with the experimental lipophilic data.
Collapse
Affiliation(s)
- Andrzej Bak
- Institute of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland
| | - Violetta Kozik
- Institute of Chemistry, University of Silesia, Szkolna 9, 40-006 Katowice, Poland
| | | | - Wojciech Baran
- Department of General and Analytical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Adam Smolinski
- Central Mining Institute, Plac Gwarków 1, 40-166 Katowice, Poland
| | - Andrzej Zięba
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland
| |
Collapse
|
3
|
Molecular Imaging of Ultrasound-Mediated Blood-Brain Barrier Disruption in a Mouse Orthotopic Glioblastoma Model. Pharmaceutics 2022; 14:pharmaceutics14102227. [PMID: 36297663 PMCID: PMC9610067 DOI: 10.3390/pharmaceutics14102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive and malignant primary brain tumor. The blood-brain barrier (BBB) limits the therapeutic options available to tackle this incurable tumor. Transient disruption of the BBB by focused ultrasound (FUS) is a promising and safe approach to increase the brain and tumor concentration of drugs administered systemically. Non-invasive, sensitive, and reliable imaging approaches are required to better understand the impact of FUS on the BBB and brain microenvironment. In this study, nuclear imaging (SPECT/CT and PET/CT) was used to quantify neuroinflammation 48 h post-FUS and estimate the influence of FUS on BBB opening and tumor growth in vivo. BBB disruptions were performed on healthy and GBM-bearing mice (U-87 MG xenograft orthotopic model). The BBB recovery kinetics were followed and quantified by [99mTc]Tc-DTPA SPECT/CT imaging at 0.5 h, 3 h and 24 h post-FUS. The absence of neuroinflammation was confirmed by [18F]FDG PET/CT imaging 48 h post-FUS. The presence of the tumor and its growth were evaluated by [68Ga]Ga-RGD2 PET/CT imaging and post-mortem histological analysis, showing that tumor growth was not influenced by FUS. In conclusion, molecular imaging can be used to evaluate the time frame for systemic treatment combined with transient BBB opening and to test its efficacy over time.
Collapse
|
4
|
Luo Z, Hu D, Gao D, Yi Z, Zheng H, Sheng Z, Liu X. High-Specificity In Vivo Tumor Imaging Using Bioorthogonal NIR-IIb Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102950. [PMID: 34617645 DOI: 10.1002/adma.202102950] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/10/2021] [Indexed: 06/13/2023]
Abstract
Lanthanide-based NIR-IIb nanoprobes are ideal for in vivo imaging. However, existing NIR-IIb nanoprobes often suffer from low tumor-targeting specificity, limiting their widespread use. Here the application of bioorthogonal nanoprobes with high tumor-targeting specificity for in vivo NIR-IIb luminescence imaging and magnetic resonance imaging (MRI) is reported. These dual-modality nanoprobes can enhance NIR-IIb emission by 20-fold and MRI signal by twofold, compared with non-bioorthogonal nanoprobes in murine subcutaneous tumors. Moreover, these bioorthogonal probes enable orthotopic brain tumor imaging. Implementation of bio-orthogonal chemistry significantly reduces the nanoprobe dose and hence cytotoxicity, providing a paradigm for real-time in vivo visualization of tumors.
Collapse
Affiliation(s)
- Zichao Luo
- Department of Chemistry and The N.1 Institute for Health, National University of Singapore, Singapore, 117543, Singapore
| | - Dehong Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS key laboratory of health informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Duyang Gao
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS key laboratory of health informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Zhigao Yi
- Department of Chemistry and The N.1 Institute for Health, National University of Singapore, Singapore, 117543, Singapore
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS key laboratory of health informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Zonghai Sheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Key Laboratory for Magnetic Resonance and Multimodality Imaging of Guangdong Province, Shenzhen Key Laboratory of Ultrasound Imaging and Therapy, CAS key laboratory of health informatics, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Xiaogang Liu
- Department of Chemistry and The N.1 Institute for Health, National University of Singapore, Singapore, 117543, Singapore
| |
Collapse
|
5
|
A natural protein based platform for the delivery of Temozolomide acid to glioma cells. Eur J Pharm Biopharm 2021; 169:297-308. [PMID: 34678408 DOI: 10.1016/j.ejpb.2021.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/17/2021] [Accepted: 10/13/2021] [Indexed: 01/13/2023]
Abstract
Glioblastoma is one of the most difficult to treat cancers with poor prognosis and survival of around one year from diagnosis. Effective treatments are desperately needed. This work aims to prepare temozolomide acid (TMZA) loaded albumin nanoparticles, for the first time, to target glioblastoma (GL261) and brain cancer stem cells (BL6). TMZA was loaded into human serum albumin nanoparticles (HSA NPs) using the desolvation method. A response surface 3-level factorial design was used to study the effect of different formulation parameters on the drug loading and particle size of NPs. The optimum conditions were found to be: 4 mg TMZA with 0.05% sodium cholate. This yielded NPs with particle size and drug loading of 111.7 nm and 5.5% respectively. The selected formula was found to have good shelf life and serum stability but with a relatively fast drug release pattern. The optimized NPs showed excellent cellular uptake with ∼ 50 and 100% of cells were positive for NP uptake after 24 h incubation with both GL261 and BL6 glioblastoma cell lines, respectively. The selected formula showed high cytotoxicity with ̴ 20% cell viability at 1 mM TMZA after 72 h incubation time. Finally, the fluorescently labelled NPs showed co-localization with the bioluminescent syngeneic BL6 intra-cranial tumour mouse model after intravenous administration.
Collapse
|
6
|
Saravanakumar K, Mariadoss AVA, Sathiyaseelan A, Wang MH. Synthesis and characterization of nano-chitosan capped gold nanoparticles with multifunctional bioactive properties. Int J Biol Macromol 2020; 165:747-757. [DOI: 10.1016/j.ijbiomac.2020.09.177] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/08/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
|
7
|
Tavassoly O, Safavi F, Tavassoly I. Heparin-binding Peptides as Novel Therapies to Stop SARS-CoV-2 Cellular Entry and Infection. Mol Pharmacol 2020; 98:612-619. [PMID: 32913137 PMCID: PMC7610036 DOI: 10.1124/molpharm.120.000098] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/27/2020] [Indexed: 01/07/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are cell surface receptors that are involved in the cellular uptake of pathologic amyloid proteins and viruses, including the novel coronavirus; severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Heparin and heparan sulfate antagonize the binding of these pathogens to HSPGs and stop their cellular internalization, but the anticoagulant effect of these agents has been limiting their use in the treatment of viral infections. Heparin-binding peptides (HBPs) are suitable nonanticoagulant agents that are capable of antagonizing binding of heparin-binding pathogens to HSPGs. Here, we review and discuss the use of HBPs as viral uptake inhibitors and will address their benefits and limitations to treat viral infections. Furthermore, we will discuss a variant of these peptides that is in the clinic and can be considered as a novel therapy in coronavirus disease 2019 (COVID-19) infection. SIGNIFICANCE STATEMENT: The need to discover treatment modalities for COVID-19 is a necessity, and therapeutic interventions such as heparin-binding peptides (HBPs), which are used for other cases, can be beneficial based on their mechanisms of actions. In this paper, we have discussed the application of HBPs as viral uptake inhibitors in COVID-19 and explained possible mechanisms of actions and the therapeutic effects.
Collapse
Affiliation(s)
- Omid Tavassoly
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (O.T.); Neuroimmunology and Neurovirology Branch, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (F.S.); and Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York (I.T.)
| | - Farinaz Safavi
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (O.T.); Neuroimmunology and Neurovirology Branch, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (F.S.); and Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York (I.T.)
| | - Iman Tavassoly
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada (O.T.); Neuroimmunology and Neurovirology Branch, National Institute of Neurologic Disorders and Stroke, National Institutes of Health, Bethesda, Maryland (F.S.); and Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York (I.T.)
| |
Collapse
|
8
|
Mahmoud BS, AlAmri AH, McConville C. Polymeric Nanoparticles for the Treatment of Malignant Gliomas. Cancers (Basel) 2020; 12:E175. [PMID: 31936740 PMCID: PMC7017235 DOI: 10.3390/cancers12010175] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/19/2019] [Accepted: 01/06/2020] [Indexed: 12/24/2022] Open
Abstract
Malignant gliomas are one of the deadliest forms of brain cancer and despite advancements in treatment, patient prognosis remains poor, with an average survival of 15 months. Treatment using conventional chemotherapy does not deliver the required drug dose to the tumour site, owing to insufficient blood brain barrier (BBB) penetration, especially by hydrophilic drugs. Additionally, low molecular weight drugs cannot achieve specific accumulation in cancerous tissues and are characterized by a short circulation half-life. Nanoparticles can be designed to cross the BBB and deliver their drugs within the brain, thus improving their effectiveness for treatment when compared to administration of the free drug. The efficacy of nanoparticles can be enhanced by surface PEGylation to allow more specificity towards tumour receptors. This review will provide an overview of the different therapeutic strategies for the treatment of malignant gliomas, risk factors entailing them as well as the latest developments for brain drug delivery. It will also address the potential of polymeric nanoparticles in the treatment of malignant gliomas, including the importance of their coating and functionalization on their ability to cross the BBB and the chemistry underlying that.
Collapse
Affiliation(s)
- Basant Salah Mahmoud
- College of Medical and Dental Sciences, School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK; (B.S.M.); or
- Hormones Department, Medical Research Division, National Research Centre, El Buhouth St., Dokki, Cairo 12622, Egypt
| | - Ali Hamod AlAmri
- College of Medical and Dental Sciences, School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK; (B.S.M.); or
- College of Pharmacy, King Khalid University, Abha 62585, Saudi Arabia
| | - Christopher McConville
- College of Medical and Dental Sciences, School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK; (B.S.M.); or
| |
Collapse
|
9
|
Derakhshankhah H, Sajadimajd S, Jafari S, Izadi Z, Sarvari S, Sharifi M, Falahati M, Moakedi F, Muganda WCA, Müller M, Raoufi M, Presley JF. Novel therapeutic strategies for Alzheimer's disease: Implications from cell-based therapy and nanotherapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 24:102149. [PMID: 31927133 DOI: 10.1016/j.nano.2020.102149] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/28/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease which leads to progressive dysfunction of cognition, memory and learning in elderly people. Common therapeutic agents are not only inadequate to suppress the progression of AD pathogenesis but also produce deleterious side effects; hence, development of alternative therapies is required to specifically suppress complications of AD. The current review provides a commentary on conventional as well as novel therapeutic approaches with an emphasis on stem cell and nano-based therapies for improvement and management of AD pathogenesis. According to our overview of the current literature, AD is a multi-factorial disorder with various pathogenic trajectories; hence, a multifunctional strategy to create effective neuroprotective agents is required to treat this disorder.
Collapse
Affiliation(s)
- Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soraya Sajadimajd
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Samira Jafari
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zhila Izadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Sarvari
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Majid Sharifi
- Department of Nanotechnology, Faculty of Advance Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advance Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Faezeh Moakedi
- Health Science Center, West Virginia University, Morgantown, USA
| | | | - Mareike Müller
- Physical Chemistry I and Research Center of Micro and Nanochemistry (Cμ), University of Siegen, Siegen, Germany
| | - Mohammad Raoufi
- Physical Chemistry I and Research Center of Micro and Nanochemistry (Cμ), University of Siegen, Siegen, Germany; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - John F Presley
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.
| |
Collapse
|
10
|
Imakita N, Kitabatake M, Ouji-Sageshima N, Hara A, Morita-Takemura S, Kasahara K, Matsukawa A, Wanaka A, Mikasa K, Ito T. Abrogated Caveolin-1 expression via histone modification enzyme Setdb2 regulates brain edema in a mouse model of influenza-associated encephalopathy. Sci Rep 2019; 9:284. [PMID: 30670717 PMCID: PMC6342998 DOI: 10.1038/s41598-018-36489-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022] Open
Abstract
Influenza-associated encephalopathy (IAE) is a serious complication that can follow influenza virus infection. Once a cytokine storm is induced during influenza virus infection, tight junction protein disruption occurs, which consequently leads to blood-brain barrier (BBB) breakdown. However, the details of IAE pathogenesis are not well understood. Here, we established a murine IAE model by administration of lipopolysaccharide following influenza virus infection. Brains from IAE model mice had significantly higher expression of type I interferons and inflammatory cytokines. In addition, the expression of Caveolin-1, one of the key proteins that correlate with protection of the BBB, was significantly lower in brains from the IAE group compared with the control group. We also found that, among 84 different histone modification enzymes, only SET domain bifurcated 2 (Setdb2), one of the histone methyltransferases that methylates the lysine 9 of histone H3, showed significantly higher expression in the IAE group compared with the control group. Furthermore, chromatin immunoprecipitation revealed that methylation of histone H3 lysine 9 was correlated with repression of the Caveolin-1 promoter region. These studies identify Caveolin-1 as a key regulator of BBB permeability in IAE and reveal that it acts through histone modification induced by Setdb2.
Collapse
Affiliation(s)
- Natsuko Imakita
- Department of Immunology, Nara Medical University, Kashihara, Nara, Japan.,Center for Infectious Diseases, Nara Medical University, Kashihara, Nara, Japan
| | | | | | - Atsushi Hara
- Department of Immunology, Nara Medical University, Kashihara, Nara, Japan
| | - Shoko Morita-Takemura
- Department of Anatomy & Neuroscience, Nara Medical University, Kashihara, Nara, Japan
| | - Kei Kasahara
- Center for Infectious Diseases, Nara Medical University, Kashihara, Nara, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Akio Wanaka
- Department of Anatomy & Neuroscience, Nara Medical University, Kashihara, Nara, Japan
| | - Keiichi Mikasa
- Center for Infectious Diseases, Nara Medical University, Kashihara, Nara, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Kashihara, Nara, Japan.
| |
Collapse
|
11
|
Gharbavi M, Amani J, Kheiri-Manjili H, Danafar H, Sharafi A. Niosome: A Promising Nanocarrier for Natural Drug Delivery through Blood-Brain Barrier. Adv Pharmacol Sci 2018; 2018:6847971. [PMID: 30651728 PMCID: PMC6311792 DOI: 10.1155/2018/6847971] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/15/2018] [Indexed: 01/25/2023] Open
Abstract
Niosomes (the nonionic surfactant vesicles), considered as novel drug delivery systems, can improve the solubility and stability of natural pharmaceutical molecules. They are established to provide targeting and controlled release of natural pharmaceutical compounds. Many factors can influence on niosome construction such as the preparation method, type and amount of surfactant, drug entrapment, temperature of lipids hydration, and the packing factor. The present review discusses about the most important features of niosomes such as their diverse structures, the different preparation approaches, characterization techniques, factors that affect their stability, their use by various routes of administration, their therapeutic applications in comparison with natural drugs, and specially the brain targeting with niosomes-ligand conjugation. It also provides recent data about the various types of ligand agents which make available active targeting drug delivery to the central neuron system. This system has an optimistic upcoming in pharmaceutical uses, mostly with the improving availability of innovative schemes to overcome blood-brain barrier and targeting the niosomes to the brain.
Collapse
Affiliation(s)
- Mahmoud Gharbavi
- School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Hossein Danafar
- School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Sharafi
- School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
12
|
Sultan D, Ye D, Heo GS, Zhang X, Luehmann H, Yue Y, Detering L, Komarov S, Taylor S, Tai YC, Rubin JB, Chen H, Liu Y. Focused Ultrasound Enabled Trans-Blood Brain Barrier Delivery of Gold Nanoclusters: Effect of Surface Charges and Quantification Using Positron Emission Tomography. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703115. [PMID: 29966035 DOI: 10.1002/smll.201703115] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 04/30/2018] [Indexed: 05/27/2023]
Abstract
Focused ultrasound (FUS) technology is reported to enhance the delivery of 64 Cu-integrated ultrasmall gold nanoclusters (64 Cu-AuNCs) across the blood-brain barrier (BBB) as measured by positron emission tomography (PET). To better define the optimal physical properties for brain delivery, 64 Cu-AuNCs with different surface charges are synthesized and characterized. In vivo biodistribution studies are performed to compare the individual organ uptake of each type of 64 Cu-AuNCs. Quantitative PET imaging post-FUS treatment shows site-targeted brain penetration, retention, and diffusion of the negative, neutral, and positive 64 Cu-AuNCs. Autoradiography is performed to compare the intrabrain distribution of these nanoclusters. PET Imaging demonstrates the effective BBB opening and successful delivery of 64 Cu-AuNCs into the brain. Of the three 64 Cu-AuNCs investigated, the neutrally charged nanostructure performs the best and is the candidate platform for future theranostic applications in neuro-oncology.
Collapse
Affiliation(s)
- Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Dezhuang Ye
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xiaohui Zhang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Lisa Detering
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sergey Komarov
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sara Taylor
- Department of Pediatrics and Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yuan-Chuan Tai
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joshua B Rubin
- Department of Pediatrics and Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hong Chen
- Department of Biomedical Engineering and Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
13
|
Rodriguez‐Otormin F, Duro‐Castano A, Conejos‐Sánchez I, Vicent MJ. Envisioning the future of polymer therapeutics for brain disorders. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1532. [DOI: 10.1002/wnan.1532] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/24/2018] [Accepted: 05/09/2018] [Indexed: 01/09/2023]
Affiliation(s)
| | - Aroa Duro‐Castano
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Valencia Spain
| | | | - María J. Vicent
- Polymer Therapeutics Laboratory Centro de Investigación Príncipe Felipe Valencia Spain
| |
Collapse
|
14
|
Agrawal M, Saraf S, Saraf S, Antimisiaris SG, Hamano N, Li SD, Chougule M, Shoyele SA, Gupta U, Ajazuddin, Alexander A. Recent advancements in the field of nanotechnology for the delivery of anti-Alzheimer drug in the brain region. Expert Opin Drug Deliv 2018; 15:589-617. [DOI: 10.1080/17425247.2018.1471058] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Mukta Agrawal
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Bhilai, Chhattisgarh, India
| | - Swarnlata Saraf
- Department of Pharmaceutics, University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh, India
| | - Shailendra Saraf
- Department of Pharmaceutics, University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh, India
- Durg University, Govt. Vasudev Vaman Patankar Girls’ P.G. College Campus, Raipur Naka, Durg, Chhattisgarh, India
| | - Sophia G. Antimisiaris
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, University of Patras, Rio, 26510, Greece
- Department of Pharmacy, FORTH/ICE-HT, Institute of Chemical Engineering, Rio, Patras, 25104, Greece
| | - Nobuhito Hamano
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British columbia V6T 1Z3, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British columbia V6T 1Z3, Canada
| | - Mahavir Chougule
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, Oxford, MS, 38677, USA
- Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS, USA
| | - Sunday A. Shoyele
- Department of Pharmaceutical Sciences, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer – 305817, India
| | - Ajazuddin
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Bhilai, Chhattisgarh, India
| | - Amit Alexander
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Bhilai, Chhattisgarh, India
| |
Collapse
|
15
|
McCully M, Sánchez-Navarro M, Teixidó M, Giralt E. Peptide Mediated Brain Delivery of Nano- and Submicroparticles: A Synergistic Approach. Curr Pharm Des 2018; 24:1366-1376. [PMID: 29205110 PMCID: PMC6110044 DOI: 10.2174/1381612824666171201115126] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 12/25/2022]
Abstract
The brain is a complex, regulated organ with a highly controlled access mechanism: The Blood-Brain Barrier (BBB). The selectivity of this barrier is a double-edged sword, being both its greatest strength and weakness. This weakness is evident when trying to target therapeutics against diseases within the brain. Diseases such as metastatic brain cancer have extremely poor prognosis due to the poor permeability of many therapeutics across the BBB. Peptides can be designed to target BBB receptors and gain access to the brain by transcytosis. These peptides (known as BBB-shuttles) can carry compounds, usually excluded from the brain, across the BBB. BBB-shuttles are limited by poor loading of therapeutics and degradation of the peptide and cargo. Likewise, nano- submicro- and microparticles can be fine-tuned to limit their degradation and with high loading of therapeutics. However, most nano- and microparticles' core materials completely lack efficient targeting, with a few selected materials able to cross the BBB passively. Combining the selectivity of peptides with the high loading potential of nano-, microparticles offers an exciting strategy to develop novel, targeted therapeutics towards many brain disorders and diseases. Nevertheless, at present the field is diverse, in both scope and nomenclature, often with competing or contradictory names. In this review, we will try to address some of these issues and evaluate the current state of peptide mediated nano,-microparticle transport to the brain, analyzing delivery vehicle type and peptide design, the two key components that must act synergistically for optimal therapeutic impact.
Collapse
Affiliation(s)
| | | | - Meritxell Teixidó
- Address correspondence to these authors at the Institute for Research in Biomedicine, Baldiri Reixac 10, 08028 Barcelona, Spain; Tel/Fax: +34 93 40 37125 0; E-mails: ;
| | - Ernest Giralt
- Address correspondence to these authors at the Institute for Research in Biomedicine, Baldiri Reixac 10, 08028 Barcelona, Spain; Tel/Fax: +34 93 40 37125 0; E-mails: ;
| |
Collapse
|
16
|
Maritim S, Coman D, Huang Y, Rao JU, Walsh JJ, Hyder F. Mapping Extracellular pH of Gliomas in Presence of Superparamagnetic Nanoparticles: Towards Imaging the Distribution of Drug-Containing Nanoparticles and Their Curative Effect on the Tumor Microenvironment. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:3849373. [PMID: 29362558 PMCID: PMC5736903 DOI: 10.1155/2017/3849373] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/25/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022]
Abstract
Since brain's microvasculature is compromised in gliomas, intravenous injection of tumor-targeting nanoparticles containing drugs (D-NPs) and superparamagnetic iron oxide (SPIO-NPs) can deliver high payloads of drugs while allowing MRI to track drug distribution. However, therapeutic effect of D-NPs remains poorly investigated because superparamagnetic fields generated by SPIO-NPs perturb conventional MRI readouts. Because extracellular pH (pHe) is a tumor hallmark, mapping pHe is critical. Brain pHe is measured by biosensor imaging of redundant deviation in shifts (BIRDS) with lanthanide agents, by detecting paramagnetically shifted resonances of nonexchangeable protons on the agent. To test the hypothesis that BIRDS-based pHe readout remains uncompromised by presence of SPIO-NPs, we mapped pHe in glioma-bearing rats before and after SPIO-NPs infusion. While SPIO-NPs accumulation in the tumor enhanced MRI contrast, the pHe inside and outside the MRI-defined tumor boundary remained unchanged after SPIO-NPs infusion, regardless of the tumor type (9L versus RG2) or agent injection method (renal ligation versus coinfusion with probenecid). These results demonstrate that we can simultaneously and noninvasively image the specific location and the healing efficacy of D-NPs, where MRI contrast from SPIO-NPs can track their distribution and BIRDS-based pHe can map their therapeutic impact.
Collapse
Affiliation(s)
- Samuel Maritim
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Daniel Coman
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Yuegao Huang
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Jyotsna U. Rao
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - John J. Walsh
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Fahmeed Hyder
- Magnetic Resonance Research Center, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| |
Collapse
|
17
|
Jiang XC, Gao JQ. Exosomes as novel bio-carriers for gene and drug delivery. Int J Pharm 2017; 521:167-175. [PMID: 28216464 DOI: 10.1016/j.ijpharm.2017.02.038] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 12/21/2022]
Abstract
Clinical treatments have stalled in certain diseases due to a lack of proper therapeutic delivery systems. Recent studies have identified exosomes for their potential use as cell-free therapies, which may provide a novel mechanism for solving this problem. Exosomes are nanoscale extracellular vesicles that can transport rich cargos of proteins, lipids, DNA, and RNA. It is increasingly recognized that exosomes play a complex role in not only the physiological conditions but also pathological ones. Accumulating evidence suggests that exosomes are of paramount importance in distant cell-cell communication because they can enter the circulation when secreted and pass through additional biological barriers. As a result, interest has exploded surrounding the functional parameters of exosomes and their potential applications as delivery vehicles for small molecule therapies. In this review, we discuss the potential of exosomes to be utilized as "natural nanoparticles" to deliver drugs and genes, and their advantages and disadvantages are compared to other delivery mechanisms.
Collapse
Affiliation(s)
- Xin-Chi Jiang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, PR China.
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, PR China.
| |
Collapse
|
18
|
Nounou MI, Adkins CE, Rubinchik E, Terrell-Hall TB, Afroz M, Vitalis T, Gabathuler R, Tian MM, Lockman PR. Anti-cancer Antibody Trastuzumab-Melanotransferrin Conjugate (BT2111) for the Treatment of Metastatic HER2+ Breast Cancer Tumors in the Brain: an In-Vivo Study. Pharm Res 2016; 33:2930-2942. [PMID: 27528392 DOI: 10.1007/s11095-016-2015-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/08/2016] [Indexed: 01/16/2023]
Abstract
PURPOSE The ability of human melanotransferrin (hMTf) to carry a therapeutic concentration of trastuzumab (BTA) in the brain after conjugation (in the form of trastuzumab-melanotransferrin conjugate, BT2111 conjugate) was investigated by measuring the reduction of the number and size of metastatic human HER2+ breast cancer tumors in a preclinical model of brain metastases of breast cancer. METHODS Human metastatic brain seeking breast cancer cells were injected in NuNu mice (n = 6-12 per group) which then developed experimental brain metastases. Drug uptake was analyzed in relation to metastasis size and blood-tumor barrier permeability. To investigate in-vivo activity against brain metastases, equimolar doses of the conjugate, and relevant controls (hMTf and BTA) in separate groups were administered biweekly after intracardiac injection of the metastatic cancer cells. RESULTS The trastuzumab-melanotransferrin conjugate (BT2111) reduced the number of preclinical human HER2+ breast cancer metastases in the brain by 68% compared to control groups. Tumors which remained after treatment were 46% smaller than the control groups. In contrast, BTA alone had no effect on reducing number of metastases, and was associated with only a minimal reduction in metastasis size. CONCLUSIONS The results suggest the novel trastuzumab-melanotransferrin conjugate (BT2111) may have utility in treating brain metastasis and validate hMTf as a potential vector for antibody transport across the Blood Brain Barrier (BBB).
Collapse
Affiliation(s)
- Mohamed Ismail Nounou
- School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, USA.,Department of Pharmaceutics, School of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Pharmaceutical Sciences, Appalachian College of Pharmacy, Oakwood, Virginia, 24631, USA
| | - Chris E Adkins
- School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, USA.,School of Pharmacy, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | | | - Tori B Terrell-Hall
- School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, USA.,School of Pharmacy, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Mohamed Afroz
- School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, USA.,School of Pharmacy, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Tim Vitalis
- biOasis Technologies Inc., Vancouver, British Columbia, Canada
| | | | - Mei Mei Tian
- biOasis Technologies Inc., Vancouver, British Columbia, Canada
| | - Paul R Lockman
- School of Pharmacy, West Virginia University Health Sciences Center, Morgantown, West Virginia, USA.
| |
Collapse
|
19
|
Cho H, Lee HY, Han M, Choi JR, Ahn S, Lee T, Chang Y, Park J. Localized Down-regulation of P-glycoprotein by Focused Ultrasound and Microbubbles induced Blood-Brain Barrier Disruption in Rat Brain. Sci Rep 2016; 6:31201. [PMID: 27510760 PMCID: PMC4980618 DOI: 10.1038/srep31201] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/13/2016] [Indexed: 01/14/2023] Open
Abstract
Multi-drug resistant efflux transporters found in Blood-Brain Barrier (BBB) acts as a functional barrier, by pumping out most of the drugs into the blood. Previous studies showed focused ultrasound (FUS) induced microbubble oscillation can disrupt the BBB by loosening the tight junctions in the brain endothelial cells; however, no study was performed to investigate its impact on the functional barrier of the BBB. In this study, the BBB in rat brains were disrupted using the MRI guided FUS and microbubbles. The immunofluorescence study evaluated the expression of the P-glycoprotein (P-gp), the most dominant multi-drug resistant protein found in the BBB. Intensity of the P-gp expression at the BBB disruption (BBBD) regions was significantly reduced (63.2 ± 18.4%) compared to the control area. The magnitude of the BBBD and the level of the P-gp down-regulation were significantly correlated. Both the immunofluorescence and histologic analysis at the BBBD regions revealed no apparent damage in the brain endothelial cells. The results demonstrate that the FUS and microbubbles can induce a localized down-regulation of P-gp expression in rat brain. The study suggests a clinically translation of this method to treat neural diseases through targeted delivery of the wide ranges of brain disorder related drugs.
Collapse
Affiliation(s)
- HongSeok Cho
- Daegu-Gyeongbuk Medical Innovation Foundation, Medical Device Development Center, Daegu, 41061, South Korea
| | - Hwa-Youn Lee
- Daegu-Gyeongbuk Medical Innovation Foundation, Medical Device Development Center, Daegu, 41061, South Korea
| | - Mun Han
- Kyungpook National University, Department of Medical &Biological Engineering, Daegu, 41566, South Korea
| | - Jong-Ryul Choi
- Daegu-Gyeongbuk Medical Innovation Foundation, Medical Device Development Center, Daegu, 41061, South Korea
| | - Sanghyun Ahn
- Daegu-Gyeongbuk Medical Innovation Foundation, Laboratory Animal Center, Daegu, 41061, South Korea
| | - Taekwan Lee
- Daegu-Gyeongbuk Medical Innovation Foundation, Laboratory Animal Center, Daegu, 41061, South Korea
| | - Yongmin Chang
- Kyungpook National University, Department of Medical &Biological Engineering, Daegu, 41566, South Korea
| | - Juyoung Park
- Daegu-Gyeongbuk Medical Innovation Foundation, Medical Device Development Center, Daegu, 41061, South Korea
| |
Collapse
|
20
|
Srimanee A, Regberg J, Hällbrink M, Vajragupta O, Langel Ü. Role of scavenger receptors in peptide-based delivery of plasmid DNA across a blood-brain barrier model. Int J Pharm 2016; 500:128-35. [PMID: 26773601 DOI: 10.1016/j.ijpharm.2016.01.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/31/2015] [Accepted: 01/05/2016] [Indexed: 01/01/2023]
Abstract
Receptor-mediated transcytosis remains a major route for drug delivery across the blood-brain barrier (BBB). PepFect 32 (PF32), a peptide-based vector modified with targeting ligand (Angiopep-2) binding to low-density lipoprotein receptor-related protein-1 (LRP-1), was previously found to be a promising vector for plasmid delivery across an in vitro model of the BBB. Cellular uptake of PF32/plasmid DNA (pDNA) complexes was speculated the internalization via LRP-1 receptor. In this study, we prove that PF32/pDNA nanocomplexes are not only transported into brain endothelial cells via LRP-1 receptor-mediated endocytosis, but also via scavenger receptor class A and B (SCARA3, SCARA5, and SR-BI)-mediated endocytosis. SCARA3, SCARA5, and SR-BI are found to be expressed in the brain endothelial cells. Inhibition of these receptors leads to a reduction of the transfection. In conclusion, this study shows that scavenger receptors also play an essential role in the cellular uptake of the PF32/pDNA nanocomplexes.
Collapse
Affiliation(s)
- Artita Srimanee
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayudhya Rd., 10400 Bangkok, Thailand; Department of Neurochemistry, The Arrhenius Laboratories for Natural Sciences, Stockholm University, SE-106 91 Stockholm, Sweden.
| | - Jakob Regberg
- Department of Neurochemistry, The Arrhenius Laboratories for Natural Sciences, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Mattias Hällbrink
- Department of Neurochemistry, The Arrhenius Laboratories for Natural Sciences, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Opa Vajragupta
- Excellent Center for Innovation in Drug Design and Discovery, Faculty of Pharmacy, Mahidol University, 447 Sri-Ayudhya Rd., 10400 Bangkok, Thailand
| | - Ülo Langel
- Department of Neurochemistry, The Arrhenius Laboratories for Natural Sciences, Stockholm University, SE-106 91 Stockholm, Sweden; Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| |
Collapse
|
21
|
In vivo Functional Evaluation of Increased Brain Delivery of the Opioid Peptide DAMGO by Glutathione-PEGylated Liposomes. Pharm Res 2015; 33:177-85. [DOI: 10.1007/s11095-015-1774-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 08/10/2015] [Indexed: 12/16/2022]
|
22
|
Abstract
Alzheimer's disease (AD), the most common form of dementia, is now representing one of the largest unmet medical needs. However, no effective treatment is now available to impede the progression of AD or delay its onset. There are two major challenges for the development of effective therapy for AD. First, the exact cause for AD onset is still unknown. Second, brain drug delivery is significantly hindered by the blood-brain barrier (BBB). In this review, we will summarize the pathological understanding about AD and the related treatments, compare BBB and its effect on brain drug delivery under normal and AD conditions and review the nanotherapeutic strategies that have been developed for AD therapy in recent years.
Collapse
|
23
|
Blood–brain barrier (BBB) toxicity and permeability assessment after L-(4-10Boronophenyl)alanine, a conventional B-containing drug for boron neutron capture therapy, using an in vitro BBB model. Brain Res 2014; 1583:34-44. [DOI: 10.1016/j.brainres.2014.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 08/07/2014] [Indexed: 12/24/2022]
|
24
|
Goldsmith M, Abramovitz L, Peer D. Precision nanomedicine in neurodegenerative diseases. ACS NANO 2014; 8:1958-65. [PMID: 24660817 DOI: 10.1021/nn501292z] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The treatment of neurodegenerative diseases remains a tremendous challenge due to the limited access of molecules across the blood-brain barrier, especially large molecules such as peptides and proteins. As a result, at most, a small percentage of a drug that is administered systemically will reach the central nervous system in its active form. Currently, research in the field focuses on developing safer and more effective approaches to deliver peptides and proteins into the central nervous system. Multiple strategies have been developed for this purpose. However, noninvasive approaches, such as nanostructured protein delivery carriers and intranasal administration, seem to be the most promising strategies for the treatment of chronic diseases, which require long-term interventions. These approaches are both target-specific and able to rapidly bypass the blood-brain barrier. In this Perspective, we detail some of these strategies and discuss some of the potential pitfalls and opportunities in this field. The next generation strategies will most likely be more cell-type-specific. Devising these strategies to target the brain may ultimately become a novel therapeutic modality to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Meir Goldsmith
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Science, Department of Material Science and Engineering, and the Center for Nanoscience and Nanotechnology, Tel Aviv University , Tel Aviv 69978, Israel
| | | | | |
Collapse
|
25
|
Yoshida Y, Hoshino S, Aisu N, Naito M, Tanimura S, Sasaki T, Takeno S, Yamashita Y. Efficacy of XELOX plus Bevacizumab in Brain Metastasis from Rectal Cancer. Case Rep Oncol 2014; 7:117-21. [PMID: 24707258 PMCID: PMC3975757 DOI: 10.1159/000360132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Brain metastasis (BM) is rare in colorectal cancer (CRC) patients. Although BM from CRC is a late-stage phenomenon with an extremely poor prognosis, some subsets of patients would benefit from a multidisciplinary management strategy. The prognosis of patients with BM from CRC was associated with the curability of the therapy for BM and the number of metastatic organs. Metastatic brain tumors are generally treated with radiotherapy because many anticancer drugs cannot cross the blood-brain barrier. Here, we present a case treated with XELOX (capecitabine and oxaliplatin) plus bevacizumab for BM from rectal cancer. To our knowledge, this is the first report of a patient who was successfully treated for BM from CRC without radiotherapy. The findings could lead to a paradigm shift in the use of chemotherapy for BM from CRC.
Collapse
Affiliation(s)
- Yoichiro Yoshida
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Seiichiro Hoshino
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Naoya Aisu
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Masayasu Naito
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Syu Tanimura
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Takamitsu Sasaki
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Shinsuke Takeno
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yuichi Yamashita
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| |
Collapse
|
26
|
Di Paolo A, Gori G, Tascini C, Danesi R, Del Tacca M. Clinical pharmacokinetics of antibacterials in cerebrospinal fluid. Clin Pharmacokinet 2014; 52:511-42. [PMID: 23605634 DOI: 10.1007/s40262-013-0062-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In the past 20 years, an increased discrepancy between new available antibacterials and the emergence of multidrug-resistant strains has been observed. This condition concerns physicians involved in the treatment of central nervous system (CNS) infections, for which clinical and microbiological success depends on the rapid achievement of bactericidal concentrations. In order to accomplish this aim, the choice of drugs is based on their disposition toward the cerebrospinal fluid (CSF), which is influenced by the physicochemical characteristics of antibacterials. A reduced distribution into CSF has been documented for beta-lactams, especially cephalosporins and carbapenems, on the basis of their hydrophilic nature. However, they represent a cornerstone of the majority of combined therapeutic schemes for their ability to achieve bactericidal concentrations, especially in the presence of inflamed meninges. The good tolerability of beta-lactams makes possible high daily dose intensities, which may be associated with increased probability of cure. Furthermore, the adoption of continuous infusion seems to be a fruitful option. Fluoroquinolones, namely moxifloxacin, and antituberculosis drugs, together with the agents such as linezolid, reach the highest CSF/plasma concentration ratio, which is greater than 0.8, and for most of these drugs it is near 1. For all drugs that are currently used for the treatment of CNS infections, the evaluation of pharmacokinetic/pharmacodynamic parameters, on the basis of dosing regimens and their time-dependent or concentration-dependent pattern of bacterial killing, remains an important aspect of clinical investigation and medical practice.
Collapse
Affiliation(s)
- Antonello Di Paolo
- Division of Pharmacology, Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | | | | | | | | |
Collapse
|
27
|
Abstract
Gelucire® 50/13, a macrogol glyceride, was used as a surfactant for the preparation and stabilization of paliperidone-loaded Capmul® GMS-50K matrix-based solid lipid nanoparticles (SLNs). The homogeneously distributed paliperidone did not affect the crystal structure of the lipid matrix in the SLNs.
Collapse
Affiliation(s)
- Sacheen Kumar
- Centre for Material Science and Engineering
- National Institute of Technology Hamirpur
- , India
| | - Jaspreet K. Randhawa
- Centre for Material Science and Engineering
- National Institute of Technology Hamirpur
- , India
| |
Collapse
|
28
|
Prokai-Tatrai K, Nguyen V, Szarka S, Konya K, Prokai L. Design and exploratory neuropharmacological evaluation of novel thyrotropin-releasing hormone analogs and their brain-targeting bioprecursor prodrugs. Pharmaceutics 2013; 5:318-28. [PMID: 24058724 PMCID: PMC3777413 DOI: 10.3390/pharmaceutics5020318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Efforts to take advantage of the beneficial activities of thyrotropin-releasing hormone (TRH) in the brain are hampered by its poor metabolic stability and lack of adequate central nervous system bioavailability. We report here novel and metabolically stable analogs that we derived from TRH by replacing its amino-terminal pyroglutamyl (pGlu) residue with pyridinium-containing moieties. Exploratory studies have shown that the resultant compounds were successfully delivered into the mouse brain after systemic administration via their bioprecursor prodrugs, where they manifested neuropharmacological responses characteristic of the endogenous parent peptide. On the other hand, the loss of potency compared to TRH in a model testing antidepressant-like effect with a simultaneous preservation of analeptic activity has been observed, when pGlu was replaced with trigonelloyl residue. This finding may indicate an opportunity for designing TRH analogs with potential selectivity towards cholinergic effects.
Collapse
Affiliation(s)
- Katalin Prokai-Tatrai
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107-2699, USA; E-Mails: (V.N.); (S.S.); (L.P.)
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107-2699, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-817-735-0617; Fax: +1-817-735-2118
| | - Vien Nguyen
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107-2699, USA; E-Mails: (V.N.); (S.S.); (L.P.)
| | - Szabolcs Szarka
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107-2699, USA; E-Mails: (V.N.); (S.S.); (L.P.)
| | - Krisztina Konya
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107-2699, USA; E-Mails: (V.N.); (S.S.); (L.P.)
| | - Laszlo Prokai
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107-2699, USA; E-Mails: (V.N.); (S.S.); (L.P.)
| |
Collapse
|
29
|
Ye D, Raghnaill MN, Bramini M, Mahon E, Åberg C, Salvati A, Dawson KA. Nanoparticle accumulation and transcytosis in brain endothelial cell layers. NANOSCALE 2013; 5:11153-65. [PMID: 24077327 DOI: 10.1039/c3nr02905k] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The blood-brain barrier (BBB) is a selective barrier, which controls and limits access to the central nervous system (CNS). The selectivity of the BBB relies on specialized characteristics of the endothelial cells that line the microvasculature, including the expression of intercellular tight junctions, which limit paracellular permeability. Several reports suggest that nanoparticles have a unique capacity to cross the BBB. However, direct evidence of nanoparticle transcytosis is difficult to obtain, and we found that typical transport studies present several limitations when applied to nanoparticles. In order to investigate the capacity of nanoparticles to access and transport across the BBB, several different nanomaterials, including silica, titania and albumin- or transferrin-conjugated gold nanoparticles of different sizes, were exposed to a human in vitro BBB model of endothelial hCMEC/D3 cells. Extensive transmission electron microscopy imaging was applied in order to describe nanoparticle endocytosis and typical intracellular localisation, as well as to look for evidence of eventual transcytosis. Our results show that all of the nanoparticles were internalised, to different extents, by the BBB model and accumulated along the endo-lysosomal pathway. Rare events suggestive of nanoparticle transcytosis were also observed for several of the tested materials.
Collapse
Affiliation(s)
- Dong Ye
- Centre for BioNano Interactions, School of Chemistry and Chemical Biology & UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | | | | | | | | | | | | |
Collapse
|
30
|
Design of brain imaging agents for positron emission tomography: do large bioconjugates provide an opportunity for in vivo brain imaging? Future Med Chem 2013; 5:1621-34. [PMID: 24047268 DOI: 10.4155/fmc.13.128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The development of brain imaging agents for positron emission tomography and other in vivo imaging modalities mostly relies on small compounds of low MW as a result of the restricted transport of larger molecules, such as peptides and proteins, across the blood–brain barrier. Besides passive transport, only a few active carrier mechanisms, such as glucose transporters and amino acid transporters, have so far been exploited to mediate the accumulation of imaging probes in the brain. An important question for the future is whether some of the abundant active carrier systems located at the blood–brain barrier can be used to shuttle potential, but non-crossing, imaging agents into the brain. What are the biological and chemical constrictions toward such bioconjugates and is it worthwhile to persue such a delivery strategy?
Collapse
|
31
|
Braccioli L, van Velthoven C, Heijnen CJ. Exosomes: a new weapon to treat the central nervous system. Mol Neurobiol 2013; 49:113-9. [PMID: 23857502 DOI: 10.1007/s12035-013-8504-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 07/01/2013] [Indexed: 12/16/2022]
Abstract
The potential of exosomes to treat central nervous system (CNS) pathologies has been recently demonstrated. These studies make way for a complete new field that aims to exploit the natural characteristics of these vesicles, considered for a long time as side products of physiological cellular pathways. Recently, however, the biological significance of exosomes has been evaluated and exosomes can now be viewed upon as new relevant functional entities for development of novel therapeutic strategies. In this review, we aim to summarize the state-of-the-art role of exosomes in the CNS and to speculate about possible future therapeutic applications of exosomes. In particular, we will speculate about the use of these vesicles as a substitute of cell-based therapies for the treatment of brain damage and review the potential of exosomes as drug delivery vehicles for the CNS.
Collapse
Affiliation(s)
- Luca Braccioli
- Laboratory for Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Lundlaan 6, 3584EA, Utrecht, The Netherlands
| | | | | |
Collapse
|
32
|
Watts RJ, Dennis MS. Bispecific antibodies for delivery into the brain. Curr Opin Chem Biol 2013; 17:393-9. [PMID: 23570979 DOI: 10.1016/j.cbpa.2013.03.023] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 02/15/2013] [Accepted: 03/18/2013] [Indexed: 01/12/2023]
Abstract
The blood-brain barrier (BBB) is a formidable obstacle preventing drug delivery to the brain, particularly for large protein therapeutics. The utilization of endogenous brain endothelial transport pathways, however, represents a promising approach to cross the cellular barrier through receptor-mediated transcytosis. Therapeutics designed to take advantage of this approach require at least two functionalities, one that facilitates transport and the other to provide therapeutic benefit, and bispecific antibodies are ideally suited for this task.
Collapse
Affiliation(s)
- Ryan J Watts
- Neurodegeneration Labs, Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, United States
| | | |
Collapse
|
33
|
Engineering a lysosomal enzyme with a derivative of receptor-binding domain of apoE enables delivery across the blood-brain barrier. Proc Natl Acad Sci U S A 2013; 110:2999-3004. [PMID: 23382178 DOI: 10.1073/pnas.1222742110] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
To realize the potential of large molecular weight substances to treat neurological disorders, novel approaches are required to surmount the blood-brain barrier (BBB). We investigated whether fusion of a receptor-binding peptide from apolipoprotein E (apoE) with a potentially therapeutic protein can bind to LDL receptors on the BBB and be transcytosed into the CNS. A lysosomal enzyme, α-L-iduronidase (IDUA), was used for biological and therapeutic evaluation in a mouse model of mucopolysaccharidosis (MPS) type I, one of the most common lysosomal storage disorders with CNS deficits. We identified two fusion candidates, IDUAe1 and IDUAe2, by in vitro screening, that exhibited desirable receptor-mediated binding, endocytosis, and transendothelial transport as well as appropriate lysosomal enzyme trafficking and biological function. Robust peripheral IDUAe1 or IDUAe2 generated by transient hepatic expression led to elevated enzyme levels in capillary-depleted, enzyme-deficient brain tissues and protein delivery into nonendothelium perivascular cells, neurons, and astrocytes within 2 d of treatment. Moreover, 5 mo after long-term delivery of moderate levels of IDUAe1 derived from maturing red blood cells, 2% to 3% of normal brain IDUA activities were obtained in MPS I mice, and IDUAe1 protein was detected in neurons and astrocytes throughout the brain. The therapeutic potential was demonstrated by normalization of brain glycosaminoglycan and β-hexosaminidase in MPS I mice 5 mo after moderate yet sustained delivery of IDUAe1. These findings provide a noninvasive and BBB-targeted procedure for the delivery of large-molecule therapeutic agents to treat neurological lysosomal storage disorders and potentially other diseases that involve the brain.
Collapse
|
34
|
Arvanitis CD, Livingstone MS, Vykhodtseva N, McDannold N. Controlled ultrasound-induced blood-brain barrier disruption using passive acoustic emissions monitoring. PLoS One 2012; 7:e45783. [PMID: 23029240 PMCID: PMC3454363 DOI: 10.1371/journal.pone.0045783] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 08/24/2012] [Indexed: 01/14/2023] Open
Abstract
The ability of ultrasonically-induced oscillations of circulating microbubbles to permeabilize vascular barriers such as the blood-brain barrier (BBB) holds great promise for noninvasive targeted drug delivery. A major issue has been a lack of control over the procedure to ensure both safe and effective treatment. Here, we evaluated the use of passively-recorded acoustic emissions as a means to achieve this control. An acoustic emissions monitoring system was constructed and integrated into a clinical transcranial MRI-guided focused ultrasound system. Recordings were analyzed using a spectroscopic method that isolates the acoustic emissions caused by the microbubbles during sonication. This analysis characterized and quantified harmonic oscillations that occur when the BBB is disrupted, and broadband emissions that occur when tissue damage occurs. After validating the system's performance in pilot studies that explored a wide range of exposure levels, the measurements were used to control the ultrasound exposure level during transcranial sonications at 104 volumes over 22 weekly sessions in four macaques. We found that increasing the exposure level until a large harmonic emissions signal was observed was an effective means to ensure BBB disruption without broadband emissions. We had a success rate of 96% in inducing BBB disruption as measured by in contrast-enhanced MRI, and we detected broadband emissions in less than 0.2% of the applied bursts. The magnitude of the harmonic emissions signals was significantly (P<0.001) larger for sonications where BBB disruption was detected, and it correlated with BBB permeabilization as indicated by the magnitude of the MRI signal enhancement after MRI contrast administration (R(2) = 0.78). Overall, the results indicate that harmonic emissions can be a used to control focused ultrasound-induced BBB disruption. These results are promising for clinical translation of this technology.
Collapse
Affiliation(s)
- Costas D Arvanitis
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | | | | | | |
Collapse
|
35
|
Yamamoto D, Iwase S, Tsubota Y, Sueoka N, Yamamoto C, Kitamura K, Odagiri H, Nagumo Y. Bevacizumab in the treatment of five patients with breast cancer and brain metastases: Japan Breast Cancer Research Network-07 trial. Onco Targets Ther 2012; 5:185-9. [PMID: 23049262 PMCID: PMC3459838 DOI: 10.2147/ott.s36515] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Brain metastases from breast cancer occur in 20%-40% of patients, and the frequency has increased over time. New radiosensitizers and cytotoxic or cytostatic agents, and innovative techniques of drug delivery are still under investigation. METHODS Five patients with brain metastases who did not respond to whole-brain radiotherapy and then received bevacizumab combined with paclitaxel were identified using our database of records between 2011 and 2012. The clinicopathological data and outcomes for these patients were then reviewed. RESULTS The median time to disease progression was 86 days. Of five patients, two (40%) achieved a partial response, two had stable disease, and one had progressive disease. In addition, one patient with brain metastases had ptosis and diplopia due to metastases of the right extraocular muscles. However, not only the brain metastases, but also the ptosis and diplopia began to disappear after 1 month of treatment. The most common treatment-related adverse events (all grades) were hypertension (60%), neuropathy (40%), and proteinuria (20%). No grade 3 toxicity was seen. No intracranial hemorrhage was observed. CONCLUSION We present five patients with breast cancer and brain metastases, with benefits from systemic chemotherapy when combined with bevacizumab.
Collapse
Affiliation(s)
- Daigo Yamamoto
- Department of Surgery, Kansai Medical University, Hirakata, Osaka ; Department of Internal Medicine, Seiko Hospital, Neyagawa, Osaka
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Vlieghe P, Khrestchatisky M. Medicinal chemistry based approaches and nanotechnology-based systems to improve CNS drug targeting and delivery. Med Res Rev 2012; 33:457-516. [PMID: 22434495 DOI: 10.1002/med.21252] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The central nervous system (CNS) is protected by various barriers, which regulate nervous tissue homeostasis and control the selective and specific uptake, efflux, and metabolism of endogenous and exogenous molecules. Among these barriers is the blood-brain barrier (BBB), a physical and physiological barrier that filters very efficiently and selectively the entry of compounds from the blood to the brain and protects nervous tissue from harmful substances and infectious agents present in the bloodstream. The BBB also prevents the entry of potential drugs. As a result, various drug targeting and delivery strategies are currently being developed to enhance the transport of drugs from the blood to the brain. Following a general introduction, we briefly overview in this review article the fundamental physiological properties of the BBB. Then, we describe current strategies to bypass the BBB (i.e., invasive methods, alternative approaches, and temporary opening) and to cross it (i.e., noninvasive approaches). This section is followed by a chapter addressing the chemical and technological solutions developed to cross the BBB. A special emphasis is given to prodrug-targeting approaches and targeted nanotechnology-based systems, two promising strategies for BBB targeting and delivery of drugs to the brain.
Collapse
Affiliation(s)
- Patrick Vlieghe
- VECT-HORUS S.A.S., Faculté de Médecine Secteur Nord, CS80011, Boulevard Pierre Dramard, 13344 Marseille Cedex 15, France.
| | | |
Collapse
|
37
|
Malcor JD, Payrot N, David M, Faucon A, Abouzid K, Jacquot G, Floquet N, Debarbieux F, Rougon G, Martinez J, Khrestchatisky M, Vlieghe P, Lisowski V. Chemical Optimization of New Ligands of the Low-Density Lipoprotein Receptor as Potential Vectors for Central Nervous System Targeting. J Med Chem 2012; 55:2227-41. [DOI: 10.1021/jm2014919] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Jean-Daniel Malcor
- Institut des Biomolécules Max-Mousseron, UMR5247 CNRS, UFR
des Sciences Pharmaceutiques et Biologiques, Universités Montpellier I et II, 15 Avenue Charles Flahault, 34093
Montpellier Cedex 5, France
| | - Nadine Payrot
- Institut des Biomolécules Max-Mousseron, UMR5247 CNRS, UFR
des Sciences Pharmaceutiques et Biologiques, Universités Montpellier I et II, 15 Avenue Charles Flahault, 34093
Montpellier Cedex 5, France
- VECT-HORUS S.A.S., Faculté de Médecine
Secteur Nord, CS80011, Boulevard Pierre Dramard, 13344 Marseille Cedex
15, France
| | - Marion David
- VECT-HORUS S.A.S., Faculté de Médecine
Secteur Nord, CS80011, Boulevard Pierre Dramard, 13344 Marseille Cedex
15, France
| | - Aude Faucon
- VECT-HORUS S.A.S., Faculté de Médecine
Secteur Nord, CS80011, Boulevard Pierre Dramard, 13344 Marseille Cedex
15, France
| | - Karima Abouzid
- VECT-HORUS S.A.S., Faculté de Médecine
Secteur Nord, CS80011, Boulevard Pierre Dramard, 13344 Marseille Cedex
15, France
| | - Guillaume Jacquot
- VECT-HORUS S.A.S., Faculté de Médecine
Secteur Nord, CS80011, Boulevard Pierre Dramard, 13344 Marseille Cedex
15, France
| | - Nicolas Floquet
- Institut des Biomolécules Max-Mousseron, UMR5247 CNRS, UFR
des Sciences Pharmaceutiques et Biologiques, Universités Montpellier I et II, 15 Avenue Charles Flahault, 34093
Montpellier Cedex 5, France
| | - Franck Debarbieux
- Aix-Marseille Univ, UMR6216, Institut de Biologie du Développement de Marseille Luminy, 13288 Marseille Cedex 9, France
- CNRS, UMR6216, Institut de Biologie du Développement de Marseille Luminy, 13288 Marseille Cedex 9, France
| | - Geneviève Rougon
- Aix-Marseille Univ, UMR6216, Institut de Biologie du Développement de Marseille Luminy, 13288 Marseille Cedex 9, France
- CNRS, UMR6216, Institut de Biologie du Développement de Marseille Luminy, 13288 Marseille Cedex 9, France
| | - Jean Martinez
- Institut des Biomolécules Max-Mousseron, UMR5247 CNRS, UFR
des Sciences Pharmaceutiques et Biologiques, Universités Montpellier I et II, 15 Avenue Charles Flahault, 34093
Montpellier Cedex 5, France
| | - Michel Khrestchatisky
- Aix-Marseille Univ, UMR7259, Laboratoire NICN, Faculté de Médecine Secteur Nord, 13344 Marseille Cedex 15, France
- CNRS, UMR7259, Laboratoire NICN, Faculté de Médecine Secteur Nord, 13344 Marseille Cedex 15, France
| | - Patrick Vlieghe
- VECT-HORUS S.A.S., Faculté de Médecine
Secteur Nord, CS80011, Boulevard Pierre Dramard, 13344 Marseille Cedex
15, France
| | - Vincent Lisowski
- Institut des Biomolécules Max-Mousseron, UMR5247 CNRS, UFR
des Sciences Pharmaceutiques et Biologiques, Universités Montpellier I et II, 15 Avenue Charles Flahault, 34093
Montpellier Cedex 5, France
| |
Collapse
|
38
|
Improved Histone Deacetylase Inhibitors as Therapeutics for the Neurodegenerative Disease Friedreich's Ataxia: A New Synthetic Route. Pharmaceuticals (Basel) 2011; 4:1578-1590. [PMID: 27721337 PMCID: PMC4060102 DOI: 10.3390/ph4121578] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 11/30/2011] [Accepted: 11/30/2011] [Indexed: 11/17/2022] Open
Abstract
Friedreich's ataxia (FRDA) is caused by transcriptional repression of the nuclear FXN gene encoding the essential mitochondrial protein frataxin. Based on the hypothesis that the acetylation state of the histone proteins is responsible for gene silencing in FRDA, previous work in our lab identified a first generation of HDAC inhibitors (pimelic o-aminobenzamides), which increase FXN mRNA in lymphocytes from FRDA patients. Importantly, these compounds also function in a FRDA mouse model to increase FXN mRNA levels in the brain and heart. While the first generation of HDAC inhibitors hold promise as potential therapeutics for FRDA, they have two potential problems: less than optimal brain penetration and metabolic instability in acidic conditions. Extensive optimization focusing on modifying the left benzene ring, linker and the right benzene ring lead to a novel class of HDAC inhibitors that have optimized pharmacological properties (increased brain penetration and acid stability) compared to the previous HDAC inhibitors. This article will describe the chemical synthesis and pharmacological properties of these new HDAC inhibitors.
Collapse
|
39
|
Efficient CRM197-mediated drug targeting to monocytes. J Control Release 2011; 158:139-47. [PMID: 21982901 DOI: 10.1016/j.jconrel.2011.09.091] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 09/21/2011] [Accepted: 09/23/2011] [Indexed: 01/04/2023]
Abstract
Efficient delivery of drugs to specific cellular reservoirs is of particular importance for therapeutics that are not able to pass cellular barriers and that may have unwanted side effects in off-target tissues. Heparin-binding epidermal growth factor (HB-EGF) is expressed on leukocytes and may be targeted for specific drug delivery using cross-reacting material (CRM)197, a non-toxic variant of diphtheria toxin and exogenous substrate for HB-EGF. We used fluorescently labeled CRM197 and CRM197-coated liposomes to investigate their potential use for drug delivery to leukocytes. We demonstrate that CRM197-guided systems are efficiently taken up by human leukocytes in vitro. CRM197 was also found to specifically target leukocytes in vivo in mice with components of the human immune system (HIS mice) and hamsters. Monocytes represent the most prominent subset of leukocytes that showed highly specific CRM197-mediated uptake. We therefore propose the application of CRM197 as a novel targeting approach in diseases that require the selective treatment of monocytes.
Collapse
|
40
|
The role of the blood-brain barrier during Venezuelan equine encephalitis virus infection. J Virol 2011; 85:10682-90. [PMID: 21849461 DOI: 10.1128/jvi.05032-11] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Venezuelan equine encephalitis (VEE) virus is a mosquito-borne alphavirus associated with sporadic outbreaks in human and equid populations in the Western Hemisphere. After the bite of an infected mosquito, the virus initiates a biphasic disease: a peripheral phase with viral replication in lymphoid and myeloid tissues, followed by a neurotropic phase with infection of central nervous system (CNS) neurons, causing neuropathology and in some cases fatal encephalitis. The mechanisms allowing VEE virus to enter the CNS are currently poorly understood. Previous data have shown that the virus gains access to the CNS by infecting olfactory sensory neurons in the nasal mucosa of mice. However, at day 5 after inoculation, the infection of the brain is multifocal, indicating that virus particles are able to cross the blood-brain barrier (BBB). To better understand the role of the BBB during VEE virus infection, we used a well-characterized mouse model system. Using VEE virus replicon particles (VRP), we modeled the early events of neuroinvasion, showing that the replication of VRP in the nasal mucosa induced the opening of the BBB, allowing peripherally administered VRP to invade the brain. Peripheral VEE virus infection was characterized by a biphasic opening of the BBB. Further, inhibition of BBB opening resulted in a delayed viral neuroinvasion and pathogenesis. Overall, these results suggest that VEE virus initially enters the CNS through the olfactory pathways and initiates viral replication in the brain, which induces the opening of the BBB, allowing a second wave of invading virus from the periphery to enter the brain.
Collapse
|
41
|
Xu S, Wang L, El-Banna M, Sohar I, Sleat DE, Lobel P. Large-volume intrathecal enzyme delivery increases survival of a mouse model of late infantile neuronal ceroid lipofuscinosis. Mol Ther 2011; 19:1842-8. [PMID: 21730969 DOI: 10.1038/mt.2011.130] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Late infantile neuronal ceroid lipofuscinosis (LINCL) is a progressive neurodegenerative lysosomal storage disorder caused by mutations in TPP1, the gene encoding the lysosomal protease tripeptidyl-peptidase (TPP1). LINCL primarily affects children, is fatal and there is no effective treatment. Administration of recombinant protein has proved effective in treatment of visceral manifestations of other lysosomal storage disorders but to date, only marginal improvement in survival has been obtained for neurological diseases. In this study, we have developed and optimized a large-volume intrathecal administration strategy to deliver therapeutic amounts of TPP1 to the central nervous system (CNS) of a mouse model of LINCL. To determine the efficacy of treatment, we have monitored survival as the primary endpoint and demonstrate that an acute treatment regimen (three consecutive daily doses started at 4 weeks of age) increases median lifespan of the LINCL mice from 16 (vehicle treated) to 23 weeks (enzyme treated). Consistent with the increase in life-span, we also observed significant reversal of pathology and improvement in neurological phenotype. These results provide a strong basis for both clinical investigation of large-volume/high-dose delivery of TPP1 to the brain via the cerebrospinal fluid (CSF) and extension of this approach towards other neurological lysosomal storage diseases.
Collapse
Affiliation(s)
- Su Xu
- Center for Advanced Biotechnology and Medicine, Piscataway, New Jersey 08854, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Malakoutikhah M, Teixidó M, Giralt E. Schleuservermittelter Transport von Wirkstoffen ins Gehirn. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201006565] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
43
|
Malakoutikhah M, Teixidó M, Giralt E. Shuttle-Mediated Drug Delivery to the Brain. Angew Chem Int Ed Engl 2011; 50:7998-8014. [DOI: 10.1002/anie.201006565] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 01/17/2011] [Indexed: 12/12/2022]
|
44
|
Malerba F, Paoletti F, Capsoni S, Cattaneo A. Intranasal delivery of therapeutic proteins for neurological diseases. Expert Opin Drug Deliv 2011; 8:1277-96. [PMID: 21619468 DOI: 10.1517/17425247.2011.588204] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Among the range of therapeutic protein candidates for new generation treatments of neurological diseases, neurotrophic factors and recombinant antibodies hold the greatest potential. However, major difficulties in their safe and effective delivery to the brain severely limit these applications. The BBB restricts the exchange of proteins between the plasma and the CNS. Moreover, therapeutic proteins often need to be selectively targeted to the brain, while minimizing their biodistribution to systemic compartments, to avoid peripheral side effects. The intranasal delivery of proteins has recently emerged as a non-invasive, safe and effective method to target proteins to the CNS, bypassing the BBB and minimizing systemic exposure. AREAS COVERED We critically summarize the main experimental and mechanistic facts about the simple and non-invasive nasal delivery approach, which provides a promising strategy and a potential solution for the severe unmet medical need of safely and effectively delivering protein therapeutics to the brain. EXPERT OPINION The intranasal route for the effective delivery of recombinant therapeutic proteins represents an emerging and promising non-invasive strategy. Future studies will achieve a detailed understanding of pharmacokinetic and mechanisms of delivery to optimize formulations and fully exploit the nose-to-brain interface in order to deliver proteins for the treatment of neurological diseases. This expanding research area will most likely produce exciting results in the near future towards new therapeutical approaches for the CNS.
Collapse
|
45
|
Cooke MJ, Wang Y, Morshead CM, Shoichet MS. Controlled epi-cortical delivery of epidermal growth factor for the stimulation of endogenous neural stem cell proliferation in stroke-injured brain. Biomaterials 2011; 32:5688-97. [PMID: 21550655 DOI: 10.1016/j.biomaterials.2011.04.032] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 04/12/2011] [Indexed: 01/06/2023]
Abstract
One of the challenges in treating central nervous system (CNS) disorders with biomolecules is achieving local delivery while minimizing invasiveness. For the treatment of stroke, stimulation of endogenous neural stem/progenitor cells (NSPCs) by growth factors is a promising strategy for tissue regeneration. Epidermal growth factor (EGF) enhances proliferation of endogenous NSPCs in the subventricular zone (SVZ) when delivered directly to the ventricles of the brain; however, this strategy is highly invasive. We designed a biomaterials-based strategy to deliver molecules directly to the brain without tissue damage. EGF or poly(ethylene glycol)-modified EGF (PEG-EGF) was dispersed in a hyaluronan and methylcellulose (HAMC) hydrogel and placed epi-cortically on both uninjured and stroke-injured mouse brains. PEG-modification decreased the rate of EGF degradation by proteases, leading to a significant increase in protein accumulation at greater tissue depths than previously shown. Consequently, EGF and PEG-EGF increased NSPC proliferation in uninjured and stroke-injured brains; and in stroke-injured brains, PEG-EGF significantly increased NSPC stimulation. Our epi-cortical delivery system is a minimally-invasive method for local delivery to the brain, providing a new paradigm for local delivery to the brain.
Collapse
Affiliation(s)
- Michael J Cooke
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON, Canada M5S 3E5
| | | | | | | |
Collapse
|
46
|
Abstract
Brain metastases are a serious obstacle in the treatment of patients with solid tumors and contribute to the morbidity and mortality of these cancers. It is speculated that the frequency of brain metastasis is increasing for several reasons, including improved systemic therapy and survival, and detection of metastases in asymptomatic patients. The lack of preclinical models that recapitulate the clinical setting and the exclusion of patients with brain metastases from most clinical trials have slowed progress. Molecular factors contributing to brain metastases are being elucidated, such as genes involved in cell adhesion, extravasation, metabolism, and cellular signaling. Furthermore, the role of the unique brain microenvironment is beginning to be explored. Although the presence and function of the blood-brain barrier in metastatic tumors is still poorly understood, it is likely that some tumor cells are protected from therapeutics by the blood-tumor barrier, creating a sanctuary site. This Review discusses what is known about the biology of brain metastases, what preclinical models are available to study the disease, and which novel therapeutic strategies are being studied in patients.
Collapse
|
47
|
Wager TT, Villalobos A, Verhoest PR, Hou X, Shaffer CL. Strategies to optimize the brain availability of central nervous system drug candidates. Expert Opin Drug Discov 2011; 6:371-81. [DOI: 10.1517/17460441.2011.564158] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
48
|
Wang CL, Guo C, Wang YQ, Zhou Y, Li Q, Ni JM, Wang R. Synthesis and antinociceptive effects of endomorphin-1 analogs with C-terminal linked by oligoarginine. Peptides 2011; 32:293-9. [PMID: 21055434 DOI: 10.1016/j.peptides.2010.10.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 10/25/2010] [Accepted: 10/25/2010] [Indexed: 10/18/2022]
Abstract
Endomorphins (EMs) cannot be delivered into the central nervous system (CNS) in sufficient quantity to elicit antinociception when given systemically because they are severely restricted by the blood-brain barrier (BBB). In the present study, we investigated herein a series of EM-1 analogs with C-terminal linked by oligoarginine in order to improve the brain delivery and antinociception after systemic administration. Indeed, all these analogs decreased the opioid receptor affinity and in vitro pharmacological activity. Moreover, analogs 4, 7-9 produced a less potent antinociceptive activity after intracerebroventricular (i.c.v.) administration, with the ED(50) values about 11- to 13-fold lower potencies than that of EM-1. Nevertheless, our results revealed that EM-1 failed to induce any significant antinociception at a dose of 50μmol/kg after subcutaneous (s.c.) administration, whereas equimolar dose of these four analogs produced a little low but significant antinociceptive effects. Naloxone (10nmol/kg, i.c.v.) significantly blocked the antinociceptive effects, indicating an opioid and central mechanism. These results demonstrated that C-terminal of EM-1 linked to oligoarginine improved the brain delivery, eliciting potent antinociception following peripheral administration.
Collapse
MESH Headings
- Analgesics, Opioid/chemical synthesis
- Analgesics, Opioid/chemistry
- Analgesics, Opioid/metabolism
- Analgesics, Opioid/pharmacology
- Animals
- Arginine/chemistry
- Brain/metabolism
- Chromatography, High Pressure Liquid
- Female
- Guinea Pigs
- Ileum/drug effects
- Male
- Mice
- Mice, Inbred Strains
- Muscle Contraction/drug effects
- Naloxone/pharmacology
- Oligopeptides/chemical synthesis
- Oligopeptides/chemistry
- Oligopeptides/metabolism
- Oligopeptides/pharmacology
- Pain/prevention & control
- Pain Measurement
- Radioligand Assay
- Rats
- Rats, Wistar
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/metabolism
- Spectrometry, Mass, Electrospray Ionization
- Vas Deferens/drug effects
Collapse
Affiliation(s)
- Chang-lin Wang
- Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou 730000, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The delivery of drugs to the CNS is hampered by the existence of the blood–brain barrier (BBB). Nowadays, medicinal chemists follow defined rules for the development of drugs able to cross the BBB. At the same time, the parameters needed in order to gain valuable estimates of brain drug delivery are well defined. Despite the limits in molecular weight that allow drugs to cross the BBB, it was shown that nanotech products, in particular properly functionalized nanoparticles, spherical particles of approximately 200 nm in diameter, are able to cross the BBB after intravenous administration and act as drug carriers for CNS. Moreover, peptides as ligands for receptors present on the brain endothelium, or able to cross the BBB and to act as carriers for CNS drug delivery in the form of conjugates with drugs, have been discovered and started to be studied as targeting moieties for nanoparticulate systems. This article will discuss the results obtained so far in the field of nanoparticle drug carriers for CNS and highlight the parameters needed in order to fully characterize these hitherto largely unknown delivery systems. Even if promising results have been obtained, more studies are needed in order to fully evaluate the clinical potential of this drug-delivery system.
Collapse
|
50
|
Meng Q, Yu M, Gu B, Li J, Liu Y, Zhan C, Xie C, Zhou J, Lu W. Myristic acid-conjugated polyethylenimine for brain-targeting delivery: in vivo and ex vivo imaging evaluation. J Drug Target 2010; 18:438-46. [PMID: 20017586 DOI: 10.3109/10611860903494229] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
To investigate the potential of myristic acid (MC) to mediate brain delivery of polyethylenimine (PEI) as a gene delivery system, a covalent conjugate (MC-PEI) of MC, and PEI was synthesized. A near-infrared fluorescence probe, IR820 was conjugated to MC-PEI to explore its in vivo distribution after intravenous (i.v.) administration in mice. The brain targeting ability of MC-PEI was evaluated by near-infrared fluorescence imaging and analyzed semiquantitatively by fluorescence intensity, respectively. Significant NIR fluorescent signal was detected in the brain 12 h after i.v. administration and further confirmed by imaging the whole brain and brain slices. Semiquantitative results from fluorescence intensity further supported the successful brain delivery of MC-PEI which led to a very significant increase ( approximately 200%) in the brain uptake after i.v. injection in comparison with unmodified PEI. The capability of MC-PEI to condense DNA was further confirmed using agarose gel retardation assay, indicating its potential for gene delivery. The significant in vivo and ex vivo results suggest that MC-PEI is a promising brain-targeting drug delivery system, especially for gene delivery.
Collapse
Affiliation(s)
- Qinggang Meng
- School of Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|