101
|
Li C, Bian X, Liu Z, Wang X, Song X, Zhao W, Liu Y, Yu Z. Effectiveness and safety of pyrotinib-based therapy in patients with HER2-positive metastatic breast cancer: A real-world retrospective study. Cancer Med 2021; 10:8352-8364. [PMID: 34672424 PMCID: PMC8633258 DOI: 10.1002/cam4.4335] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/22/2021] [Accepted: 09/20/2021] [Indexed: 11/07/2022] Open
Abstract
The previous studies had demonstrated the promising effectiveness and acceptable safety of pyrotinib in patients with HER2‐positive metastatic breast cancer. We aimed to investigate the real‐world data of pyrotinib in complex clinical practice and complement the findings of clinical trials. Two hundred and eighteen patients were included for effectiveness analysis. A total of 62.0% had received two or more lines of systematic therapy, and 95.4% had been exposed to prior anti‐HER2 therapy, with 95.4% receiving trastuzumab, 5.0% receiving pertuzumab, and 40.8% receiving lapatinib. The median progression‐free survival (PFS) was 9.3 months and the objective response rate (ORR) was 44.0%. Patients treated with pyrotinib‐based therapy as first, second, or later line had a median PFS of 15.0, 10.3, and 6.8 months, respectively. Patients treated with pyrotinib and trastuzumab received significant benefit in terms of median PFS compared with pyrotinib alone (10.7 (9.1–12.3) vs. 8.8 (8.1–9.5), p = 0.016). Patients pretreated with lapatinib had a median PFS of 6.9 months. The median PFS time was 7.0 months in patients with brain metastasis. Multivariate Cox regression analyses showed that lines of pyrotinib‐based therapy (1 vs. 2 vs. ≥3), prior treatment with lapatinib, and combination treatments with trastuzumab proved to be independent predictors of PFS. Two hundred and forty‐eight patients were included in the safety analysis, and the results showed that the toxicity of pyrotinib was tolerable, with the most common grade 3/4 adverse event being diarrhea (19.8%). Pyrotinib‐based therapy demonstrated promising efficacy and tolerable toxicity in first‐, second‐, and later‐line treatments and in lapatinib‐treated patients. The combination of pyrotinib and trastuzumab showed advantages in PFS, even for patients resisting trastuzumab. Pyrotinib‐based therapy could be the preferred choice for brain metastasis patients, especially when combined with brain radiotherapy.
Collapse
Affiliation(s)
- Chao Li
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaoli Bian
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Breast Surgery, Heze Municipal Hospital, Heze, China
| | - Zhaoyun Liu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xinzhao Wang
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiang Song
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Zhao
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yansong Liu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhiyong Yu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
102
|
Bang S, Song JK, Shin SW, Lee KH. Human serum albumin fusion protein as therapeutics for targeting amyloid beta in Alzheimer's diseases. Neurosci Lett 2021; 767:136298. [PMID: 34673147 DOI: 10.1016/j.neulet.2021.136298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) is characterized by amyloid beta (Aβ) plaques and neurofibrillary tangles. AD drug development has been limited due to the presence of the blood-brain barrier (BBB), which prevents efficient uptake of therapeutics into the brain. To solve this problem, we used trans-activator of transcription (TAT)-transducing domain and added the human serum albumin (HSA) carrier to increase the half-life of the drug within the body. In addition, we included the protein of interest for lowering Aβ deposition and/or neurofibrillary tangles. We made HSA fusion protein (designated AL04) which contains Cystatin C (CysC) as core mechanism of action moiety in the construct containing tandem repeat TAT (dTAT). After purification of 80KDa AL04, we investigate the therapeutic potential of AL04 in vitro and AD mouse model Tg2576. We evaluated the permeability of AL04 through the BBB using a cell-basedhuman BBB model and show that dTAT plays a role in facilitating the delivery of 80 kDa protein. We found out that AL04 attenuates Aβ-induced neurotoxicity in PC12 cells. In Tg2576 mice brain, Aβ plaques were dramatically reduced in AL04 treated mice. These data suggest that BBB-crossing albumin fusion protein AL04 with CysC active moiety can be a disease modifying treatment for AD.
Collapse
|
103
|
Zheng M, Du Q, Wang X, Zhou Y, Li J, Xia X, Lu Y, Yin J, Zou Y, Park JB, Shi B. Tuning the Elasticity of Polymersomes for Brain Tumor Targeting. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102001. [PMID: 34423581 PMCID: PMC8529491 DOI: 10.1002/advs.202102001] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/13/2021] [Indexed: 05/27/2023]
Abstract
Nanoformulations show great potential for delivering drugs to treat brain tumors. However, how the mechanical properties of nanoformulations affect their ultimate brain destination is still unknown. Here, a library of membrane-crosslinked polymersomes with different elasticity are synthesized to investigate their ability to effectively target brain tumors. Crosslinked polymersomes with identical particle size, zeta potential and shape are assessed, but their elasticity is varied depending on the rigidity of incorporated crosslinkers. Benzyl and oxyethylene containing crosslinkers demonstrate higher and lower Young's modulus, respectively. Interestingly, stiff polymersomes exert superior brain tumor cell uptake, excellent in vitro blood brain barrier (BBB) and tumor penetration but relatively shorter blood circulation time than their soft counterparts. These results together affect the in vivo performance for which rigid polymersomes exerting higher brain tumor accumulation in an orthotopic glioblastoma (GBM) tumor model. The results demonstrate the crucial role of nanoformulation elasticity for brain-tumor targeting and will be useful for the design of future brain targeting drug delivery systems for the treatment of brain disease.
Collapse
Affiliation(s)
- Meng Zheng
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
| | - Qiuli Du
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
| | - Xin Wang
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
| | - Yuan Zhou
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
| | - Jia Li
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
| | - Xue Xia
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
| | - Yiqing Lu
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
- School of EngineeringFaculty of Science and EngineeringMacquarie UniversitySydneyNSW2109Australia
| | - Jinlong Yin
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
| | - Yan Zou
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
- Department of Biomedical SciencesFaculty of Medicine & Health SciencesMacquarie UniversitySydneyNSW2109Australia
| | - Jong Bae Park
- Department of Cancer Biomedical ScienceGraduate School of Cancer Science and PolicyNational Cancer CenterGoyang10408South Korea
| | - Bingyang Shi
- Henan and Macquarie University Joint Centre for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifeng475004China
- Henan Key Laboratory of Brain Targeted Bio‐nanomedicineSchool of Life Sciences & School of PharmacyHenan UniversityKaifeng475004China
- Department of Biomedical SciencesFaculty of Medicine & Health SciencesMacquarie UniversitySydneyNSW2109Australia
| |
Collapse
|
104
|
Qi YL, Wang HR, Chen LL, Guo L, Cao YY, Yang YS, Duan YT, Zhu HL. Recent advances in reaction-based fluorescent probes for the detection of central nervous system-related pathologies in vivo. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214068] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
105
|
Fang Z, Chen L, Moser MAJ, Zhang W, Qin Z, Zhang B. Electroporation-Based Therapy for Brain Tumors: A Review. J Biomech Eng 2021; 143:100802. [PMID: 33991087 DOI: 10.1115/1.4051184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 12/21/2022]
Abstract
Electroporation-based therapy (EBT), as a high-voltage-pulse technology has been prevalent with favorable clinical outcomes in the treatment of various solid tumors. This review paper aims to promote the clinical translation of EBT for brain tumors. First, we briefly introduced the mechanism of pore formation in a cell membrane activated by external electric fields using a single cell model. Then, we summarized and discussed the current in vitro and in vivo preclinical studies, in terms of (1) the safety and effectiveness of EBT for brain tumors in animal models, and (2) the blood-brain barrier (BBB) disruption induced by EBT. Two therapeutic effects could be achieved in EBT for brain tumors simultaneously, i.e., the tumor ablation induced by irreversible electroporation (IRE) and transient BBB disruption induced by reversible electroporation (RE). The BBB disruption could potentially improve the uptake of antitumor drugs thereby enhancing brain tumor treatment. The challenges that hinder the application of EBT in the treatment of human brain tumors are discussed in the review paper as well.
Collapse
Affiliation(s)
- Zheng Fang
- Energy-Based Tumor Ablation Laboratory, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Lingchao Chen
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Michael A J Moser
- Department of Surgery, University of Saskatchewan, Saskatoon SK S7N 5A9, Canada
| | - Wenjun Zhang
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon SK S7N 5A9, Canada
| | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Bing Zhang
- Energy-Based Tumor Ablation Laboratory, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| |
Collapse
|
106
|
Nanotheranostic agents for neurodegenerative diseases. Emerg Top Life Sci 2021; 4:645-675. [PMID: 33320185 DOI: 10.1042/etls20190141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases (NDDs), including Alzheimer's disease (AD) and Parkinson's disease (PD), affect the ageing population worldwide and while severely impairing the quality of life of millions, they also cause a massive economic burden to countries with progressively ageing populations. Parallel with the search for biomarkers for early detection and prediction, the pursuit for therapeutic approaches has become growingly intensive in recent years. Various prospective therapeutic approaches have been explored with an emphasis on early prevention and protection, including, but not limited to, gene therapy, stem cell therapy, immunotherapy and radiotherapy. Many pharmacological interventions have proved to be promising novel avenues, but successful applications are often hampered by the poor delivery of the therapeutics across the blood-brain-barrier (BBB). To overcome this challenge, nanoparticle (NP)-mediated drug delivery has been considered as a promising option, as NP-based drug delivery systems can be functionalized to target specific cell surface receptors and to achieve controlled and long-term release of therapeutics to the target tissue. The usefulness of NPs for loading and delivering of drugs has been extensively studied in the context of NDDs, and their biological efficacy has been demonstrated in numerous preclinical animal models. Efforts have also been made towards the development of NPs which can be used for targeting the BBB and various cell types in the brain. The main focus of this review is to briefly discuss the advantages of functionalized NPs as promising theranostic agents for the diagnosis and therapy of NDDs. We also summarize the results of diverse studies that specifically investigated the usage of different NPs for the treatment of NDDs, with a specific emphasis on AD and PD, and the associated pathophysiological changes. Finally, we offer perspectives on the existing challenges of using NPs as theranostic agents and possible futuristic approaches to improve them.
Collapse
|
107
|
An J, Chen B, Tian D, Guo Y, Yan Y, Yang H. Regulation of Neurogenesis and Neuronal Differentiation by Natural Compounds. Curr Stem Cell Res Ther 2021; 17:756-771. [PMID: 34493197 DOI: 10.2174/1574888x16666210907141447] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/15/2021] [Accepted: 07/28/2021] [Indexed: 11/22/2022]
Abstract
Neuronal damage or degeneration is the main feature of neurological diseases. Regulation of neurogenesis and neuronal differentiation is important in developing therapies to promote neuronal regeneration or synaptic network reconstruction. Neurogenesis is a multistage process in which neurons are generated and integrated into existing neuronal circuits. Neuronal differentiation is extremely complex because it can occur in different cell types and can be caused by a variety of inducers. Recently, natural compounds that induce neurogenesis and neuronal differentiation have attracted extensive attention. In this paper, the potential neural induction effects of medicinal plant-derived natural compounds on neural stem/progenitor cells (NS/PCs), the cultured neuronal cells, and mesenchymal stem cells (MSCs) are reviewed. The natural compounds that are efficacious in inducing neurogenesis and neuronal differentiation include phenolic acids, polyphenols, flavonoids, glucosides, alkaloids, terpenoids, quinones, coumarins, and others. They exert neural induction effects by regulating signal factors and cell-specific genes involved in the process of neurogenesis and neuronal differentiation, including specific proteins (β-tubulin III, MAP-2, tau, nestin, neurofilaments, GFAP, GAP-43, NSE), related genes and proteins (STAT3, Hes1, Mash1, NeuroD1, notch, cyclin D1, SIRT1, reggie-1), transcription factors (CREB, Nkx-2.5, Ngn1), neurotrophins (BDNF, NGF, NT-3) and signaling pathways (JAK/STAT, Wnt/β-catenin, MAPK, PI3K/Akt, GSK-3β/β-catenin, Ca2+/CaMKII/ATF1, Nrf2/HO-1, BMP). The natural compounds with neural induction effects are of great value for neuronal regenerative medicine and provide promising prevention and treatment strategies for neurological diseases.
Collapse
Affiliation(s)
- Jing An
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Bo Chen
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Ding Tian
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Yunshan Guo
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Yuzhu Yan
- Clinical Lab, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| | - Hao Yang
- Translational Medicine Centre, Honghui Hospital, Xi'an Jiaotong University, Xi'an. China
| |
Collapse
|
108
|
Nsairat H, Khater D, Odeh F, Al-Adaileh F, Al-Taher S, Jaber AM, Alshaer W, Al Bawab A, Mubarak MS. Lipid nanostructures for targeting brain cancer. Heliyon 2021; 7:e07994. [PMID: 34632135 PMCID: PMC8488847 DOI: 10.1016/j.heliyon.2021.e07994] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/28/2021] [Accepted: 09/09/2021] [Indexed: 01/02/2023] Open
Abstract
Advancements in both material science and bionanotechnology are transforming the health care sector. To this end, nanoparticles are increasingly used to improve diagnosis, monitoring, and therapy. Huge research is being carried out to improve the design, efficiency, and performance of these nanoparticles. Nanoparticles are also considered as a major area of research and development to meet the essential requirements for use in nanomedicine where safety, compatibility, biodegradability, biodistribution, stability, and effectiveness are requirements towards the desired application. In this regard, lipids have been used in pharmaceuticals and medical formulations for a long time. The present work focuses on the use of lipid nanostructures to combat brain tumors. In addition, this review summarizes the literature pertaining to solid lipid nanoparticles (SLN) and nanostructured lipid carriers (LNC), methods of preparation and characterization, developments achieved to overcome blood brain barrier (BBB), and modifications used to increase their effectiveness.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Dima Khater
- Department of Chemistry, Faculty of Arts and Science, Applied Science Private University, Amman 11931, Jordan
| | - Fadwa Odeh
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
| | - Fedaa Al-Adaileh
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
| | - Suma Al-Taher
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
| | - Areej M. Jaber
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Abeer Al Bawab
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
- Hamdi Mango Center for Scientific Research, The University of Jordan, Amman 11942, Jordan
| | | |
Collapse
|
109
|
Elmowafy M, Al-Sanea MM. Nanostructured lipid carriers (NLCs) as drug delivery platform: Advances in formulation and delivery strategies. Saudi Pharm J 2021; 29:999-1012. [PMID: 34588846 PMCID: PMC8463508 DOI: 10.1016/j.jsps.2021.07.015] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
NLCs have provoked the incessant impulsion for the development of safe and valuable drug delivery systems owing to their exceptional physicochemical and then biocompatible characteristics. Throughout the earlier period, a lot of studies recounting NLCs based formulations have been noticeably increased. They are binary system which contains both solid and liquid lipids aiming to produce less ordered lipidic core. Their constituents particularly influence the physicochemical properties and effectiveness of the final product. NLCs can be fabricated by different techniques which are classified according to consumed energy. More utilization NLCs is essential due to overcome barriers surrounded by the technological procedure of lipid-based nanocarriers' formulation and increased information of the core mechanisms of their transport via various routes of administration. They can be used in different applications and by different routes such as oral, cutaneous, ocular and pulmonary. This review article seeks to present an overview on the existing situation of the art of NLCs for future clinics through exposition of their applications which shall foster their lucid use. The reported records evidently demonstrate the promise of NLCs for innovate therapeutic applications in the future.
Collapse
Affiliation(s)
- Mohammed Elmowafy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka P.O. Box 2014, Saudi Arabia
- Department of Pharmaceutics and Ind. Pharmacy, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Mohammad M. Al-Sanea
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72341, Aljouf Province, Saudi Arabia
| |
Collapse
|
110
|
Zhang L, Sun H, Zhao J, Lee J, Ee Low L, Gong L, Chen Y, Wang N, Zhu C, Lin P, Liang Z, Wei M, Ling D, Li F. Dynamic nanoassemblies for imaging and therapy of neurological disorders. Adv Drug Deliv Rev 2021; 175:113832. [PMID: 34146626 DOI: 10.1016/j.addr.2021.113832] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/07/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023]
Abstract
The past decades have witnessed an increased incidence of neurological disorders (NDs) such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, ischemic stroke, and epilepsy, which significantly lower patients' life quality and increase the economic and social burden. Recently, nanomedicines composed of imaging and/or therapeutic agents have been explored to diagnose and/or treat NDs due to their enhanced bioavailability, blood-brain barrier (BBB) permeability, and targeting capacity. Intriguingly, dynamic nanoassemblies self-assembled from functional nanoparticles to simultaneously interfere with multiple pathogenic substances and pathological changes, have been regarded as one of the foremost candidates to improve the diagnostic and therapeutic efficacy of NDs. To help readers better understand this emerging field, in this review, the pathogenic mechanism of different types of NDs is briefly introduced, then the functional nanoparticles used as building blocks in the construction of dynamic nanoassemblies for NDs theranostics are summarized. Furthermore, dynamic nanoassemblies that can actively cross the BBB to target brain lesions, sensitively and efficiently diagnose or treat NDs, and effectively promote neuroregeneration are highlighted. Finally, we conclude with our perspectives on the future development in this field.
Collapse
|
111
|
Gabay M, Weizman A, Zeineh N, Kahana M, Obeid F, Allon N, Gavish M. Liposomal Carrier Conjugated to APP-Derived Peptide for Brain Cancer Treatment. Cell Mol Neurobiol 2021; 41:1019-1029. [PMID: 33025416 DOI: 10.1007/s10571-020-00969-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/22/2020] [Indexed: 01/17/2023]
Abstract
Brain tumors are hard to treat with the currently available therapy. The major obstacle in the treatment of brain tumors is the lack of therapeutic strategies capable to penetrate the blood-brain barrier (BBB). The BBB is an endothelial interface that separates the brain from the circulatory blood system and prevents the exposure of the central nervous system (CNS) to circulating toxins and potentially harmful compounds. Unfortunately, the BBB prevents also the penetration of therapeutic compounds into the brain. We present here a drug-delivery liposomal carrier, conjugated to a peptide inserted in the liposomal membrane, which is putatively recognized by BBB transporters. The peptide is a short sequence of 5 amino acids (RERMS) present in the amyloid precursor protein (APP). This APP-targeted liposomal system was designed specifically for transporting compounds with anti-cancer activity via the BBB into the brain in an effective manner. This drug-delivery liposomal carrier loaded with the anti-cancer compounds temozolomide (TMZ), curcumin, and doxorubicin crossed the BBB in an in vitro model as well as in vivo (mice model). In the in vitro model, the targeted liposomes crossed the BBB model fourfold higher than the non-targeted liposomes. Labeled targeted liposomes penetrated the brain in vivo 35% more than non-targeted liposomes. Treatment of mice that underwent intracranial injection of human U87 glioblastoma, with the targeted liposomes loaded with the three tested anti-cancer agents, delayed the tumor growth and prolonged the mice survival in a range of 45% -70%. It appears that the targeted liposomal drug-delivery system enables better therapeutic efficacy in a SCID mouse model of glioblastoma compared to the corresponding non-targeted liposomes and the free compounds.
Collapse
Affiliation(s)
- Martin Gabay
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Abraham Weizman
- Research Unit, Geha Mental Health Center and the Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, 4910002, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Nidal Zeineh
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Meygal Kahana
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Fadi Obeid
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Nahum Allon
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel
| | - Moshe Gavish
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, 31096, Haifa, Israel.
| |
Collapse
|
112
|
Fayazi N, Sheykhhasan M, Soleimani Asl S, Najafi R. Stem Cell-Derived Exosomes: a New Strategy of Neurodegenerative Disease Treatment. Mol Neurobiol 2021; 58:3494-3514. [PMID: 33745116 PMCID: PMC7981389 DOI: 10.1007/s12035-021-02324-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
Short-term symptomatic treatment and dose-dependent side effects of pharmacological treatment for neurodegenerative diseases have forced the medical community to seek an effective treatment for this serious global health threat. Therapeutic potential of stem cell for treatment of neurodegenerative disorders was identified in 1980 when fetal nerve tissue was used to treat Parkinson's disease (PD). Then, extensive studies have been conducted to develop this treatment strategy for neurological disease therapy. Today, stem cells and their secretion are well-known as a therapeutic environment for the treatment of neurodegenerative diseases. This new paradigm has demonstrated special characteristics related to this treatment, including neuroprotective and neurodegeneration, remyelination, reduction of neural inflammation, and recovery of function after induced injury. However, the exact mechanism of stem cells in repairing nerve damage is not yet clear; exosomes derived from them, an important part of their secretion, are introduced as responsible for an important part of such effects. Numerous studies over the past few decades have evaluated the therapeutic potential of exosomes in the treatment of various neurological diseases. In this review, after recalling the features and therapeutic history, we will discuss the latest stem cell-derived exosome-based therapies for these diseases.
Collapse
Affiliation(s)
- Nashmin Fayazi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sara Soleimani Asl
- Anatomy Department, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
113
|
Li L, Chen J, Ming Y, Li B, Fu R, Duan D, Li Z, Ni R, Wang X, Zhou Y, Zhang L. The Application of Peptides in Glioma: a Novel Tool for Therapy. Curr Pharm Biotechnol 2021; 23:620-633. [PMID: 34182908 DOI: 10.2174/1389201022666210628114042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glioma is the most aggressive and lethal tumor of the central nervous system. Owing to the cellular heterogeneity, the invasiveness, and blood-brain barrier (BBB), current therapeutic approaches, such as chemotherapy and radiotherapy, are poorly to obtain great anti-tumor efficacy. However, peptides, a novel type of therapeutic agent, displayed excellent ability in the tumor, which becomes a new molecule for glioma treatment. METHOD We review the current knowledge on peptides for the treatment of glioma through a PubMed-based literature search. RESULTS In the treatment of glioma, peptides can be used as (i) decoration on the surface of the delivery system, facilitating the distribution and accumulation of the anti-tumor drug in the target site;(ii) anti-tumor active molecules, inhibiting the growth of glioma and reducing solid tumor volume; (iii) immune-stimulating factor, and activating immune cells in the tumor microenvironment or recruiting immune cells to the tumor for breaking out the immunosuppression by glioma cells. CONCLUSION The application of peptides has revolutionized the treatment of glioma, which is based on targeting, penetrating, anti-tumor activities, and immunostimulatory. Moreover, better outcomes have been discovered in combining different kinds of peptides rather than a single one. Until now, more and more preclinical studies have been developed with multifarious peptides, which show promising results in vitro or vivo with the model of glioma.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Jianhong Chen
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Yue Ming
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Ruoqiu Fu
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Dongyu Duan
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Ziwei Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Rui Ni
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianfeng Wang
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Yueling Zhou
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Lin Zhang
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
114
|
Peng Y, Lu J, Li R, Zhao Y, Hai L, Guo L, Wu Y. Glucose and Triphenylphosphonium Co-Modified Redox-Sensitive Liposomes to Synergistically Treat Glioma with Doxorubicin and Lonidamine. ACS APPLIED MATERIALS & INTERFACES 2021; 13:26682-26693. [PMID: 34061501 DOI: 10.1021/acsami.1c02404] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Glioma is one of the most lethal and complex tumors, and thus, an effective drug delivery system must selectively target the tumor sites and release its cargos in a controlled manner. For the first time, we combined chemotherapeutic agent doxorubicin (DOX) and chemosensitizer lonidamine (LND) to synergistically treat glioma. We also designed and prepared multitargeted redox-sensitive liposomes (Lip-SPG) co-modified with glucose and triphenylphosphonium (TPP) to effectively deliver DOX and LND for anti-glioma therapy. The anti-glioma evaluation shows that DOX and LND have a synergistic effect and Lip-SPG could further enhance their cooperation. In vitro, Lip-SPG could increase the cellular uptake and mitochondrial uptake on bEnd.3 cells and C6 cells with multitargeting ability on the brain, tumor, and mitochondria mediated by glucose and TPP. Lip-SPG can also escape from lysosomes before entering the mitochondria. The anti-glioma efficacy in vitro shows that Lip-SPG can inhibit tumor cell proliferation and induce apoptosis. In addition, Lip-SPG have a remarkable interference to mitochondria, such as reducing intracellular ATP production, inducing ROS generation, and promoting mitochondrial membrane potential depolarization. Furthermore, in vivo, the introduction of PEGylation via glutathione-sensitive disulfide bonds endows Lip-SPG with favorable pharmacokinetic properties, brain targeting ability, low toxicity to normal tissues, and great anti-glioma efficacy with the survival time extended from 19 to 39 days. In conclusion, Lip-SPG are an effective delivery system for synergistically treating glioma with DOX and LND.
Collapse
Affiliation(s)
- Yao Peng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jiaqi Lu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ru Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yi Zhao
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Li Hai
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Li Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yong Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
115
|
Kouhi A, Pachipulusu V, Kapenstein T, Hu P, Epstein AL, Khawli LA. Brain Disposition of Antibody-Based Therapeutics: Dogma, Approaches and Perspectives. Int J Mol Sci 2021; 22:ijms22126442. [PMID: 34208575 PMCID: PMC8235515 DOI: 10.3390/ijms22126442] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
Due to their high specificity, monoclonal antibodies have been widely investigated for their application in drug delivery to the central nervous system (CNS) for the treatment of neurological diseases such as stroke, Alzheimer’s, and Parkinson’s disease. Research in the past few decades has revealed that one of the biggest challenges in the development of antibodies for drug delivery to the CNS is the presence of blood–brain barrier (BBB), which acts to restrict drug delivery and contributes to the limited uptake (0.1–0.2% of injected dose) of circulating antibodies into the brain. This article reviews the various methods currently used for antibody delivery to the CNS at the preclinical stage of development and the underlying mechanisms of BBB penetration. It also describes efforts to improve or modulate the physicochemical and biochemical properties of antibodies (e.g., charge, Fc receptor binding affinity, and target affinity), to adapt their pharmacokinetics (PK), and to influence their distribution and disposition into the brain. Finally, a distinction is made between approaches that seek to modify BBB permeability and those that use a physiological approach or antibody engineering to increase uptake in the CNS. Although there are currently inherent difficulties in developing safe and efficacious antibodies that will cross the BBB, the future prospects of brain-targeted delivery of antibody-based agents are believed to be excellent.
Collapse
|
116
|
Novel brain-targeting 3-n-butylphthalide prodrugs for ischemic stroke treatment. J Control Release 2021; 335:498-514. [PMID: 34087248 DOI: 10.1016/j.jconrel.2021.05.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022]
Abstract
Currently, ischemic stroke is the leading cause of disability and death worldwide, and the performance of corresponding drugs is often unsatisfactory owing to the complex pathological processes and the impediment of the blood-brain barrier (BBB). Here, we employed various tertiary amino groups, including different linear, cyclic, and bimolecular drug structures, to modify 3-n-butylphthalide (NBP), a natural product used for ischemic stroke treatment, which has poor bioavailability, to generate a series of six prodrugs. These prodrugs showed significantly improved solubility and cellular uptake, which were primarily driven by putative pyrilamine cationic transporters. They also displayed more efficient brain delivery in vivo, reaching as high as 21.5-fold brain accumulation increase compared with NBP, leading to much higher bioavailability and stronger therapeutic effects. The toxicity of these molecules is also lower or similar to that of unmodified NBP. We showed that the tertiary amino group-modified NBP prodrugs are effective and safe for treating ischemic stroke with significantly enhanced druggability; hence, they have potential for further clinical development.
Collapse
|
117
|
Ruan S, Zhou Y, Jiang X, Gao H. Rethinking CRITID Procedure of Brain Targeting Drug Delivery: Circulation, Blood Brain Barrier Recognition, Intracellular Transport, Diseased Cell Targeting, Internalization, and Drug Release. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004025. [PMID: 33977060 PMCID: PMC8097396 DOI: 10.1002/advs.202004025] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/03/2020] [Indexed: 05/06/2023]
Abstract
The past decades have witnessed great progress in nanoparticle (NP)-based brain-targeting drug delivery systems, while their therapeutic potentials are yet to be fully exploited given that the majority of them are lost during the delivery process. Rational design of brain-targeting drug delivery systems requires a deep understanding of the entire delivery process along with the issues that they may encounter. Herein, this review first analyzes the typical delivery process of a systemically administrated NPs-based brain-targeting drug delivery system and proposes a six-step CRITID delivery cascade: circulation in systemic blood, recognizing receptor on blood-brain barrier (BBB), intracellular transport, diseased cell targeting after entering into parenchyma, internalization by diseased cells, and finally intracellular drug release. By dissecting the entire delivery process into six steps, this review seeks to provide a deep understanding of the issues that may restrict the delivery efficiency of brain-targeting drug delivery systems as well as the specific requirements that may guarantee minimal loss at each step. Currently developed strategies used for troubleshooting these issues are reviewed and some state-of-the-art design features meeting these requirements are highlighted. The CRITID delivery cascade can serve as a guideline for designing more efficient and specific brain-targeting drug delivery systems.
Collapse
Affiliation(s)
- Shaobo Ruan
- Key laboratory of Drug Targeting and Drug Delivery Systems of the Education MinistrySichuan Engineering Laboratory for Plant‐sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
- Department of PharmaceuticsCollege of PharmacyUniversity of FloridaGainesvilleFlorida32610USA
| | - Yang Zhou
- Key laboratory of Drug Targeting and Drug Delivery Systems of the Education MinistrySichuan Engineering Laboratory for Plant‐sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
| | - Xinguo Jiang
- Key laboratory of Smart Drug DeliveryMinistry of EducationSchool of PharmacyFudan UniversityShanghai201203China
| | - Huile Gao
- Key laboratory of Drug Targeting and Drug Delivery Systems of the Education MinistrySichuan Engineering Laboratory for Plant‐sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of PharmacySichuan UniversityChengdu610041China
| |
Collapse
|
118
|
Blood-brain barrier opening by intracarotid artery hyperosmolar mannitol induces sterile inflammatory and innate immune responses. Proc Natl Acad Sci U S A 2021; 118:2021915118. [PMID: 33906946 DOI: 10.1073/pnas.2021915118] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracarotid arterial hyperosmolar mannitol (ICAHM) blood-brain barrier disruption (BBBD) is effective and safe for delivery of therapeutics for central nervous system malignancies. ICAHM osmotically alters endothelial cells and tight junction integrity to achieve BBBD. However, occurrence of neuroinflammation following hemispheric BBBD by ICAHM remains unknown. Temporal proteomic changes in rat brains following ICAHM included increased damage-associated molecular patterns, cytokines, chemokines, trophic factors, and cell adhesion molecules, indicative of a sterile inflammatory response (SIR). Proteomic changes occurred within 5 min of ICAHM infusion and returned to baseline by 96 h. Transcriptomic analyses following ICAHM BBBD further supported an SIR. Immunohistochemistry revealed activated astrocytes, microglia, and macrophages. Moreover, proinflammatory proteins were elevated in serum, and proteomic and histological findings from the contralateral hemisphere demonstrated a less pronounced SIR, suggesting neuroinflammation beyond regions of ICAHM infusion. Collectively, these results demonstrate ICAHM induces a transient SIR that could potentially be harnessed for neuroimmunomodulation.
Collapse
|
119
|
Development of a Blood-Brain Barrier Permeability Assay Using Human Induced Pluripotent Stem Cell Derived Brain Endothelial Cells. Methods Mol Biol 2021; 2454:397-410. [PMID: 33881753 DOI: 10.1007/7651_2021_393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The development of translational and predictive models in vitro for assessing blood-brain barrier (BBB) delivery has become an important requirement in preclinical testing of CNS-targeting therapeutics. Here we describe a directed monolayer differentiation strategy to generate a population of brain endothelial-like cells (BECs) from human induced pluripotent stem cell (iPSC) with robust BBB properties. To generate BBB permeability assays, the BECs are seeded as a monolayer on a semipermeable Transwell insert placed inside a companion plate to generate a two-compartment Transwell model. The BECs provide a BBB-like separation between the luminal (blood) and abluminal (brain) compartments to assess BBB permeability of CNS-targeting therapeutics.
Collapse
|
120
|
Crossing the blood-brain barrier: A review on drug delivery strategies using colloidal carrier systems. Neurochem Int 2021; 147:105017. [PMID: 33887377 DOI: 10.1016/j.neuint.2021.105017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/27/2021] [Accepted: 03/06/2021] [Indexed: 02/05/2023]
Abstract
The blood-brain barrier represents the major challenge for delivering drugs to the central nervous system (CNS). It separates the blood circulation from the brain tissue, thereby protecting the CNS and maintaining its ion homeostasis. Unfortunately, most drugs are not able to cross this barrier in vivo despite promising in vitro results. One approach to solve this problem is the delivery of drugs via surface modified nanocarrier systems. This review will give an overview on currently tested systems, mainly liposomes and solid nanoparticles and inform about new developments.
Collapse
|
121
|
Zhang G, Jiang B, Wu C, Liu Y, He Y, Huang X, Chen W, Xi K, Guo H, Zhao X, Jia X. Thin platelet-like COF nanocomposites for blood brain barrier transport and inhibition of brain metastasis from renal cancer. J Mater Chem B 2021; 8:4475-4488. [PMID: 32365151 DOI: 10.1039/d0tb00724b] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Effective treatment of brain metastases is hindered by the blood-brain barrier (BBB) and the rapid development of resistance to drug therapy. Moreover, the clinical application of general formulations is hampered by biological barriers and biological elimination. To tackle this challenge, we report a feasible approach for the assembly of polymer-covalent organic framework (COF) nanocomposites into 150 nm thin platelets as a drug delivery vehicle for enhanced retention in brain tumours. Using intravital imaging, we demonstrate that these polymer-COF nanocomposites are able to traverse the BBB in mice and achieve direct tumour accumulation in intracranial orthotopic models of brain metastasis from renal cancer (BMRC). These nanocomposites can target brain tumour cells and respond to tumour microenvironmental characteristics, including acidic and redox conditions. Intracranial tumour acidity triggers the breakdown of the nanoassemblies to polymer-COF nanocomposites due to the presence of borate bonds. Furthermore, in vivo studies on the nanocomposites showed enhanced brain tumour-targeting efficiency and therapeutic effects compared to those of free-drug dosing. Mice treated with drug-loaded polymer-COF nanocomposites also show protection from systemic drug toxicity and improved survival, demonstrating the preclinical potential of this nanoscale platform to deliver novel combination therapies to BMRC and other central nervous system (CNS) tumours.
Collapse
Affiliation(s)
- Guiyang Zhang
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China. and Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Bo Jiang
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, P. R. China.
| | - Chunyong Wu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Yanfeng Liu
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China.
| | - Yidan He
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Huang
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China.
| | - Wei Chen
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, P. R. China.
| | - Kai Xi
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China.
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, P. R. China.
| | - Xiaozhi Zhao
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, P. R. China.
| | - Xudong Jia
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China.
| |
Collapse
|
122
|
Delivery of Therapeutic Agents to the Central Nervous System and the Promise of Extracellular Vesicles. Pharmaceutics 2021; 13:pharmaceutics13040492. [PMID: 33916841 PMCID: PMC8067091 DOI: 10.3390/pharmaceutics13040492] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
The central nervous system (CNS) is surrounded by the blood–brain barrier (BBB), a semipermeable border of endothelial cells that prevents pathogens, solutes and most molecules from non-selectively crossing into the CNS. Thus, the BBB acts to protect the CNS from potentially deleterious insults. Unfortunately, the BBB also frequently presents a significant barrier to therapies, impeding passage of drugs and biologicals to target cells within the CNS. This review provides an overview of different approaches to deliver therapeutics across the BBB, with an emphasis in extracellular vesicles as delivery vehicles to the CNS.
Collapse
|
123
|
Anthony DP, Hegde M, Shetty SS, Rafic T, Mutalik S, Rao BSS. Targeting receptor-ligand chemistry for drug delivery across blood-brain barrier in brain diseases. Life Sci 2021; 274:119326. [PMID: 33711385 DOI: 10.1016/j.lfs.2021.119326] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/09/2021] [Accepted: 03/03/2021] [Indexed: 12/18/2022]
Abstract
The blood-brain barrier (BBB) is composed of a layer of endothelial cells that is interspersed with a series of tight junctions and characterized by the absence of fenestrations. The permeability of this barrier is controlled by junctions such as tight junctions and adherent junctions as well as several cells such as astrocytes, pericytes, vascular endothelial cells, neurons, microglia, and efflux transporters with relatively enhanced expression. It plays a major role in maintaining homeostasis in the brain and exerts a protective regulatory control on the influx and efflux of molecules. However, it proves to be a challenge for drug delivery strategies that target brain diseases like Dementia, Parkinson's Disease, Alzheimer's Disease, Brain Cancer or Stroke, Huntington's Disease, Lou Gehrig's Disease, etc. Conventional modes of drug delivery are invasive and have been known to contribute to a "leaky BBB", recent studies have highlighted the efficiency and relative safety of receptor-mediated drug delivery. Several receptors are exhibited on the BBB, and actively participate in nutrient uptake, and recognize specific ligands that modulate the process of endocytosis. The strategy employed in receptor-mediated drug delivery exploits this process of "tricking" the receptors into internalizing ligands that are conjugated to carrier systems like liposomes, nanoparticles, monoclonal antibodies, enzymes etc. These in turn are modified with drug molecules, therefore leading to delivery to desired target cells in brain tissue. This review comprehensively explores each of those receptors that can be modified to serve such purposes as well as the currently employed strategies that have led to increased cellular uptake and transport efficiency.
Collapse
Affiliation(s)
- Danielle Paige Anthony
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Manasa Hegde
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shreya S Shetty
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Thasneema Rafic
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - B S Satish Rao
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
124
|
Gasca-Salas C, Fernández-Rodríguez B, Pineda-Pardo JA, Rodríguez-Rojas R, Obeso I, Hernández-Fernández F, Del Álamo M, Mata D, Guida P, Ordás-Bandera C, Montero-Roblas JI, Martínez-Fernández R, Foffani G, Rachmilevitch I, Obeso JA. Blood-brain barrier opening with focused ultrasound in Parkinson's disease dementia. Nat Commun 2021; 12:779. [PMID: 33536430 PMCID: PMC7859400 DOI: 10.1038/s41467-021-21022-9] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022] Open
Abstract
MR-guided focused ultrasound (MRgFUS), in combination with intravenous microbubble administration, has been applied for focal temporary BBB opening in patients with neurodegenerative disorders and brain tumors. MRgFUS could become a therapeutic tool for drug delivery of putative neurorestorative therapies. Treatment for Parkinson’s disease with dementia (PDD) is an important unmet need. We initiated a prospective, single-arm, non-randomized, proof-of-concept, safety and feasibility phase I clinical trial (NCT03608553), which is still in progress. The primary outcomes of the study were to demonstrate the safety, feasibility and reversibility of BBB disruption in PDD, targeting the right parieto-occipito-temporal cortex where cortical pathology is foremost in this clinical state. Changes in β-amyloid burden, brain metabolism after treatments and neuropsychological assessments, were analyzed as exploratory measurements. Five patients were recruited from October 2018 until May 2019, and received two treatment sessions separated by 2–3 weeks. The results are set out in a descriptive manner. Overall, this procedure was feasible and reversible with no serious clinical or radiological side effects. We report BBB opening in the parieto-occipito-temporal junction in 8/10 treatments in 5 patients as demonstrated by gadolinium enhancement. In all cases the procedures were uneventful and no side effects were encountered associated with BBB opening. From pre- to post-treatment, mild cognitive improvement was observed, and no major changes were detected in amyloid or fluorodeoxyglucose PET. MRgFUS-BBB opening in PDD is thus safe, reversible, and can be performed repeatedly. This study provides encouragement for the concept of BBB opening for drug delivery to treat dementia in PD and other neurodegenerative disorders. Blood brain barrier (BBB) opening is being investigated as a therapeutic approach for neurodegenerative diseases. Here, the authors report the results of a phase I trial to evaluate the feasibility and safety of BBB opening of the right parieto-occipito-temporal cortex in Parkinson´s disease with dementia.
Collapse
Affiliation(s)
- Carmen Gasca-Salas
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Beatriz Fernández-Rodríguez
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain
| | - José A Pineda-Pardo
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Rafael Rodríguez-Rojas
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Ignacio Obeso
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Frida Hernández-Fernández
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Department of Nursing, Universidad Europea de Madrid. Faculty of Biomedical and Health Sciences, Villaviciosa de Odón, Madrid, Spain
| | - Marta Del Álamo
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - David Mata
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Pasqualina Guida
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | | | | | - Raúl Martínez-Fernández
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Guglielmo Foffani
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.,Hospital Nacional de Parapléjicos, Toledo, Spain
| | | | - José A Obeso
- HM CINAC, Fundación HM Hospitales de Madrid, University Hospital HM Puerta del Sur. CEU-San Pablo University, Móstoles, Madrid, Spain. .,Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| |
Collapse
|
125
|
Choudhari M, Hejmady S, Narayan Saha R, Damle S, Singhvi G, Alexander A, Kesharwani P, Kumar Dubey S. Evolving new-age strategies to transport therapeutics across the blood-brain-barrier. Int J Pharm 2021; 599:120351. [PMID: 33545286 DOI: 10.1016/j.ijpharm.2021.120351] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/24/2021] [Accepted: 01/30/2021] [Indexed: 12/25/2022]
Abstract
A basic understanding of the blood-brain barrier (BBB) is essential for the novel advancements in targeting drugs specific to the brain. Neoplasm compromising the internal structure of BBB that results in impaired vasculature is called as blood tumor barrier (BTB). Besides, the BBB serves as a chief hindrance to the passage of a drug into the brain parenchyma. The small and hydrophilic drugs majorly display an absence of desired molecular characteristics required to cross the BBB. Furthermore, all classes of biologics have failed in the clinical trials of brain diseases over the past years since these biologics are large molecules that do not cross the BBB. Also, new strategies have been discovered that use the Trojan horse technology with the re-engineered biologics for BBB transport. Thus, this review delivers information about the different grades of tumors (I-IV) i.e. examples of BBB/BTB heterogenicity along with the different mechanisms for transporting the therapeutics into the brain tumors by crossing BBB. This review also provides insights into the emerging approaches of peptide delivery and the non-invasive and brain-specific molecular Trojan horse targeting technologies. Also, the several challenges in the clinical development of BBB penetrating IgG fusion protein have been discussed.
Collapse
Affiliation(s)
- Manisha Choudhari
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Siddhanth Hejmady
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Ranendra Narayan Saha
- Birla Institute of Technology and Science, Pilani, Dubai Campus, United Arab Emirates
| | - Shantanu Damle
- Colorcon Asia Pvt. Ltd., Verna Industrial Estate, Verna 403722, Goa, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Amit Alexander
- National Institute of Pharmaceutical Education and Research (NIPER GUWAHATI), Department of Pharmaceutical Technology (Formulations), Department of Pharmaceuticals, Ministry of Chemical and Fertilizers, Government of India, Sila Village, Nizsundarighopa, Changsari, Kamrup (R), Guwahati, Assam 781101, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India; R&D Healthcare Division Emami Ltd., 13, BT Road, Belgharia, Kolkata 700056, India.
| |
Collapse
|
126
|
Kumbhar SA, Kokare CR, Shrivastava B, Gorain B, Choudhury H. Antipsychotic Potential and Safety Profile of TPGS-Based Mucoadhesive Aripiprazole Nanoemulsion: Development and Optimization for Nose-To-Brain Delivery. J Pharm Sci 2021; 110:1761-1778. [PMID: 33515583 DOI: 10.1016/j.xphs.2021.01.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022]
Abstract
Delivering therapeutics to the brain using conventional dosage forms is always a challenge, thus the present study was aimed to formulate mucoadhesive nanoemulsion (MNE) of aripiprazole (ARP) for intranasal delivery to transport the drug directly to the brain. Therefore, a TPGS based ARP-MNE was formulated and optimized using the Box-Behnken statistical design. The improved in vitro release profile of the formulation was in agreement to enhanced ex vivo permeation through sheep mucous membranes with a maximum rate of permeation co-efficient (62.87 cm h-1 × 103) and flux (31.43 μg cm-2.h-1). The pharmacokinetic profile following single-dose administration showed the maximum concentration of drug in the brain (Cmax) of 15.19 ± 2.51 μg mL-1 and Tmax of 1 h in animals with ARP-MNE as compared to 10.57 ± 1.88 μg mL-1 and 1 h, and 2.52 ± 0.38 μg mL-1 and 3 h upon intranasal and intravenous administration of ARP-NE, respectively. Further, higher values of % drug targeting efficiency (96.9%) and % drug targeting potential (89.73%) of ARP-MNE through intranasal administration were investigated. The studies in Wistar rats showed no existence of extrapyramidal symptoms through the catalepsy test and forelimb retraction results. No ex vivo ciliotoxicity on nasal mucosa reflects the safety of the components and delivery tool. Further, findings on locomotor activity and hind-limb retraction test in ARP-MNE treated animals established its antipsychotic efficacy. Thus, it can be inferred that the developed ARP-MNE could effectively be explored as brain delivery cargo in the effective treatment of schizophrenia without producing any toxic manifestation.
Collapse
Affiliation(s)
- Santosh Ashok Kumbhar
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur 302 017, Rajasthan, India; Department of Pharmaceutics, STES's Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411 041, Maharashtra, India
| | - Chandrakant R Kokare
- Department of Pharmaceutics, STES's Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411 041, Maharashtra, India
| | - Birendra Shrivastava
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur 302 017, Rajasthan, India.
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Science, Taylor's University, Subang Jaya, Selangor, Malaysia; Centre for Drug Delivery and Molecular Pharmacology, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia; Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, Bukit jalil, 57000 Kuala Lumpur, Malaysia.
| |
Collapse
|
127
|
Zhou X, Smith QR, Liu X. Brain penetrating peptides and peptide-drug conjugates to overcome the blood-brain barrier and target CNS diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1695. [PMID: 33470550 DOI: 10.1002/wnan.1695] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Nearly one in six people worldwide suffer from disorders of the central nervous system (CNS). There is an urgent need for effective strategies to improve the success rates in CNS drug discovery and development. The lack of effective technologies for delivering drugs and genes to the brain due to the blood-brain barrier (BBB), a structural barrier that effectively blocks most neurotherapeutic agents from reaching the brain, has posed a formidable hurdle for CNS drug development. Brain-homing and brain-penetrating molecular transport vectors, such as brain permeable peptides or BBB shuttle peptides, have shown promise in overcoming the BBB and ferrying the drug molecules to the brain. The BBB shuttle peptides are discovered by phage display technology or derived from natural neurotropic proteins or certain viruses and harness the receptor-mediated transcytosis molecular machinery for crossing the BBB. Brain permeable peptide-drug conjugates (PDCs), composed of BBB shuttle peptides, linkers, and drug molecules, have emerged as a promising CNS drug delivery system by taking advantage of the endogenous transcytosis mechanism and tricking the brain into allowing these bioactive molecules to pass the BBB. Here, we examine the latest development of brain-penetrating peptide shuttles and brain-permeable PDCs as molecular vectors to deliver small molecule drug payloads across the BBB to reach brain parenchyma. Emerging knowledge of the contribution of the peptides and their specific receptors expressed on the brain endothelial cells, choice of drug payloads, the design of PDCs, brain entry mechanisms, and delivery efficiency to the brain are highlighted. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Quentin R Smith
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| |
Collapse
|
128
|
Elliott RO, He M. Unlocking the Power of Exosomes for Crossing Biological Barriers in Drug Delivery. Pharmaceutics 2021; 13:pharmaceutics13010122. [PMID: 33477972 PMCID: PMC7835896 DOI: 10.3390/pharmaceutics13010122] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/25/2022] Open
Abstract
Since the 2013 Nobel Prize was awarded for the discovery of vesicle trafficking, a subgroup of nanovesicles called exosomes has been driving the research field to a new regime for understanding cellular communication. This exosome-dominated traffic control system has increased understanding of many diseases, including cancer metastasis, diabetes, and HIV. In addition to the important diagnostic role, exosomes are particularly attractive for drug delivery, due to their distinctive properties in cellular information transfer and uptake. Compared to viral and non-viral synthetic systems, the natural, cell-derived exosomes exhibit intrinsic payload and bioavailability. Most importantly, exosomes easily cross biological barriers, obstacles that continue to challenge other drug delivery nanoparticle systems. Recent emerging studies have shown numerous critical roles of exosomes in many biological barriers, including the blood–brain barrier (BBB), blood–cerebrospinal fluid barrier (BCSFB), blood–lymph barrier (BlyB), blood–air barrier (BAB), stromal barrier (SB), blood–labyrinth barrier (BLaB), blood–retinal barrier (BRB), and placental barrier (PB), which opens exciting new possibilities for using exosomes as the delivery platform. However, the systematic reviews summarizing such discoveries are still limited. This review covers state-of-the-art exosome research on crossing several important biological barriers with a focus on the current, accepted models used to explain the mechanisms of barrier crossing, including tight junctions. The potential to design and engineer exosomes to enhance delivery efficacy, leading to future applications in precision medicine and immunotherapy, is discussed.
Collapse
Affiliation(s)
- Rebekah Omarkhail Elliott
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA;
| | - Mei He
- Department of Chemical and Petroleum Engineering, Bioengineering Program, University of Kansas, Lawrence, KS 66045, USA;
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
- Correspondence:
| |
Collapse
|
129
|
Santi M, Finamore F, Cecchettini A, Santorelli FM, Doccini S, Rocchiccioli S, Signore G. Protein Delivery by Peptide-Based Stealth Liposomes: A Biomolecular Insight into Enzyme Replacement Therapy. Mol Pharm 2020; 17:4510-4521. [PMID: 33112630 DOI: 10.1021/acs.molpharmaceut.0c00615] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Infantile neural ceroid lipofuscinosis (INCL) is a lysosomal storage disorder characterized by mutations in the CLN1 gene that leads to lack of the lysosomal enzyme palmitoyl-protein thioesterase-1 (PPT1), which causes the progressive death of cortical neurons. Enzyme replacement therapy (ERT) is one of the most promising treatments, but its translation toward a clinical use is hampered by the need to deliver the enzyme to the central nervous system and a more detailed understanding of its capability to restore physiologic conditions at the biochemical and protein level, beyond the simple regulation of enzymatic activity. Targeted nanoparticles can promote protein delivery to the central nervous system and affect biological pathways inside cells. Here, we describe an innovative peptide-based stealth nanoparticle that inhibits serum protein adsorption exploiting transferrin-driven internalization to convey the PPT1 enzyme to transferrin receptor-mediated pathways (endocytosis in this work, or transcytosis, in perspective, in vivo). These enzyme-loaded nanoparticles were able to restore stable levels of enzymatic activity in CLN1 patient's fibroblasts, comparable with the free enzyme, demonstrating that delivery after encapsulation in the nanocarrier does not alter uptake or intracellular trafficking. We also investigate, for the first time, dysregulated pathways of proteome and palmitoylome and their alteration upon enzyme delivery. Our nanoparticles were able of halving palmitoylated protein levels restoring conditions similar to the normal cells. From proteomic analysis, we also highlighted the reduction of the different groups of proteins after treatments with the free or encapsulated enzyme. In conclusion, our system is able to deliver the enzyme to a model of CLN1 disease restoring normal conditions in cells. Investigation of molecular details of pathologic state and enzyme-based correction reveals dysregulated pathways with unprecedented details for CLN1. Finally, we unveil for the first time the dysregulation landscape of palmitoylome and proteome in primary patient-derived fibroblasts and their modifications in response to enzyme administration. These findings will provide a guideline for the validation of future therapeutic strategies based on enzyme replacement therapy or acting at different metabolic levels.
Collapse
Affiliation(s)
- Melissa Santi
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Pisa 56127, Italy.,NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Pisa 56127, Italy
| | | | | | | | | | | | - Giovanni Signore
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Pisa 56127, Italy.,Fondazione Pisana per la Scienza, Pisa 56017, Italy
| |
Collapse
|
130
|
Chen J, Wang Y. Personalized dynamic transport of magnetic nanorobots inside the brain vasculature. NANOTECHNOLOGY 2020; 31:495706. [PMID: 33016261 DOI: 10.1088/1361-6528/abb392] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Delivering specific bioactive agents with sufficient bioavailability to the targeted brain area across blood brain barrier remains a big challenge. Magnetically driven nanorobots have demonstrated their potential for controlled drug delivery. However, the dynamic transport of these nanorobots inside each individual's brain vasculature is not yet well studied. Addressing this is a critical step forward to controlled drug delivery for non-invasive brain therapeutics. In this paper, we develop an analytical model describing the personalized dynamic transport of spherical magnetic nanorobots inside the brain vasculature reconstructed from the patient's angiography images. By inverting the transporting process, we first design the patient-specific transport path based on the reconstructed vascular model, and then calculate the magnetic force required to drive these nanorobots from the analytical model. Also, a finite element model is created to simulate the inverse design process, which implies that the delivery efficiency of these magnetically driven nanorobots to the targeted brain area can be increased by 20% and almost 95% nanorobots arrive at the desired vessel walls. In the end, a simplified brain vascular model is printed using PolyJet 3D 750 to demonstrate the dynamic transport of these nanorobots toward the targeted site. The proposed theoretical modeling, numerical simulation and experimental validation lay solid foundation toward non-invasive brain therapeutics with maximal accuracy and minimal side effects.
Collapse
Affiliation(s)
- Jingfan Chen
- J. Mike Walker' 66 Department of Mechanical Engineering, Texas A&M University, College Station, United States of America
| | - Ya Wang
- J. Mike Walker' 66 Department of Mechanical Engineering, Texas A&M University, College Station, United States of America
| |
Collapse
|
131
|
Abstract
Ischemic stroke, which is caused by a sudden clot in the blood vessels, may cause severe brain tissue damage and has become a leading cause of death globally. Currently, thrombolysis is the gold standard primary treatment of ischemic stroke in clinics. However, the short therapeutic window of opportunity limits thrombolysis utility. Secondary cerebral damage caused by stroke is also an urgent problem. In this review, we discuss the present methods of treating ischemic stroke in clinics and their limitations. Various new drug delivery strategies targeting ischemic stroke lesions have also been summarized, including pharmaceutical methods, diagnostic approaches and other routes. These strategies could change the pharmacokinetic behavior, improve targeted delivery or minimize side effects. A better understanding of the novel approaches utilized to facilitate drug delivery in ischemic stroke would improve outcomes.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| | - Rong Yan
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| | - Jingjing Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, P. R. China
| |
Collapse
|
132
|
Hanes J, Dobakova E, Majerova P. Brain Drug Delivery: Overcoming the Blood-brain Barrier to Treat Tauopathies. Curr Pharm Des 2020; 26:1448-1465. [PMID: 32178609 DOI: 10.2174/1381612826666200316130128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/10/2020] [Indexed: 02/06/2023]
Abstract
Tauopathies are neurodegenerative disorders characterized by the deposition of abnormal tau protein in the brain. The application of potentially effective therapeutics for their successful treatment is hampered by the presence of a naturally occurring brain protection layer called the blood-brain barrier (BBB). BBB represents one of the biggest challenges in the development of therapeutics for central nervous system (CNS) disorders, where sufficient BBB penetration is inevitable. BBB is a heavily restricting barrier regulating the movement of molecules, ions, and cells between the blood and the CNS to secure proper neuronal function and protect the CNS from dangerous substances and processes. Yet, these natural functions possessed by BBB represent a great hurdle for brain drug delivery. This review is concentrated on summarizing the available methods and approaches for effective therapeutics' delivery through the BBB to treat neurodegenerative disorders with a focus on tauopathies. It describes the traditional approaches but also new nanotechnology strategies emerging with advanced medical techniques. Their limitations and benefits are discussed.
Collapse
Affiliation(s)
- Jozef Hanes
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska cesta 9, 845 10 Bratislava, Slovakia
| | - Eva Dobakova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska cesta 9, 845 10 Bratislava, Slovakia
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Centre of Excellence for Alzheimer's Disease and Related Disorders, Dubravska cesta 9, 845 10 Bratislava, Slovakia
| |
Collapse
|
133
|
Baskin J, Jeon JE, Lewis SJG. Nanoparticles for drug delivery in Parkinson's disease. J Neurol 2020; 268:1981-1994. [PMID: 33141248 DOI: 10.1007/s00415-020-10291-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022]
Abstract
Although effective symptomatic treatments for Parkinson's disease (PD) have been available for some time, efficient and well-controlled drug delivery to the brain has proven to be challenging. The emergence of nanotechnology has created new opportunities not only for improving the pharmacokinetics of conventional therapies but also for developing novel treatment approaches and disease modifying therapies. Several exciting strategies including drug carrier nanoparticles targeting specific intracellular pathways and structural reconformation of tangled proteins as well as introducing reprogramming genes have already shown promise and are likely to deliver more tailored approaches to the treatment of PD in the future. This paper reviews the role of nanoparticles in PD including a discussion of both their composition and functional capacity as well as their potential to deliver better therapeutic agents.
Collapse
Affiliation(s)
- Jonathan Baskin
- Parkinson's Disease Research Clinic, Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia.
| | - June Evelyn Jeon
- Parkinson's Disease Research Clinic, Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Simon J G Lewis
- Parkinson's Disease Research Clinic, Brain and Mind Centre, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
134
|
Wang Z, Du J, Lachance BB, Mascarenhas C, He J, Jia X. Intracerebroventricular Administration of hNSCs Improves Neurological Recovery after Cardiac Arrest in Rats. Stem Cell Rev Rep 2020; 17:923-937. [PMID: 33140234 DOI: 10.1007/s12015-020-10067-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2020] [Indexed: 12/15/2022]
Abstract
Irreversible brain injury and neurological dysfunction induced by cardiac arrest (CA) have long been a clinical challenge due to lack of effective therapeutic interventions to reverse neuronal loss and prevent secondary reperfusion injury. The neuronal regenerative potential of neural stem cells (NSCs) provides a possible solution to this clinical deficit. We investigated the neuronal recovery potential of human neural stem cells (hNSCs) via intracerebroventricular (ICV) xenotransplantation after CA in rats and the effects of transplanted NSCs on the proliferation and migration of endogenous NSCs. Outcome measures included neurological functional recovery measured by neurological deficit score (NDS), electrophysiologic analysis of EEG, and assessment of proliferation and migration at the cellular level and the Wnt/β-catenin pathway at the molecular level. Neurological functional assessment based on aggregate neurological deficit score (NDS) showed better recovery of function after hNSCs therapy (P < 0.05). Tracking of stem cells' proliferation with Ki67 antibody suggested that the NSCs group had more prominent proliferation compared to control group (number of Ki67+ cells, Control VS. NSC: 89.0 ± 31.6 VS. 352.7 ± 97.3, P < 0.05). In addition, cell migration tracked by Dcx antibody showed more Dcx + cells migrated to the far distance zone from SVZ in the treatment group (P < 0.05). Further immunofluorescence staining confirmed that the expression of the Wnt signaling pathway protein (β-catenin) was upregulated in the NSC group (P < 0.05). ICV delivery of hNSCs promotes endogenous NSC proliferation and migration and ultimately enhances neuronal survival and neurological functional recovery. Wnt/β-catenin pathway may be involved in the initiation and maintenance of this enhancement.Graphical abstract.
Collapse
Affiliation(s)
- Zhuoran Wang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 43007, China.,Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MSTF Building 823, Baltimore, MD, 21201, USA
| | - Jian Du
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MSTF Building 823, Baltimore, MD, 21201, USA
| | - Brittany Bolduc Lachance
- Program in Trauma, Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Conrad Mascarenhas
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MSTF Building 823, Baltimore, MD, 21201, USA
| | - Junyun He
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MSTF Building 823, Baltimore, MD, 21201, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, 10 South Pine Street, MSTF Building 823, Baltimore, MD, 21201, USA. .,Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA. .,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA. .,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
135
|
Recent progress in therapeutic drug delivery systems for treatment of traumatic CNS injuries. Future Med Chem 2020; 12:1759-1778. [PMID: 33028091 DOI: 10.4155/fmc-2020-0178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Most therapeutics for the treatment of traumatic central nervous system injuries, such as traumatic brain injury and spinal cord injury, encounter various obstacles in reaching the target tissue and exerting pharmacological effects, including physiological barriers like the blood-brain barrier and blood-spinal cord barrier, instability rapid elimination from the injured tissue or cerebrospinal fluid and off-target toxicity. For central nervous system delivery, nano- and microdrug delivery systems are regarded as the most suitable and promising carriers. In this review, the pathophysiology and biomarkers of traumatic central nervous system injuries (traumatic brain injury and spinal cord injury) are introduced. Furthermore, various drug delivery systems, novel combinatorial therapies and advanced therapies for the treatment of traumatic brain injury and spinal cord injury are emphasized.
Collapse
|
136
|
Rana I, Khan N, Ansari MM, Shah FA, Din FU, Sarwar S, Imran M, Qureshi OS, Choi HI, Lee CH, Kim JK, Zeb A. Solid lipid nanoparticles-mediated enhanced antidepressant activity of duloxetine in lipopolysaccharide-induced depressive model. Colloids Surf B Biointerfaces 2020; 194:111209. [DOI: 10.1016/j.colsurfb.2020.111209] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022]
|
137
|
Schlachet I, Moshe Halamish H, Sosnik A. Mixed Amphiphilic Polymeric Nanoparticles of Chitosan, Poly(vinyl alcohol) and Poly(methyl methacrylate) for Intranasal Drug Delivery: A Preliminary In Vivo Study. Molecules 2020; 25:molecules25194496. [PMID: 33008001 PMCID: PMC7582691 DOI: 10.3390/molecules25194496] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 01/10/2023] Open
Abstract
Intranasal (i.n.) administration became an alternative strategy to bypass the blood-brain barrier and improve drug bioavailability in the brain. The main goal of this work was to preliminarily study the biodistribution of mixed amphiphilic mucoadhesive nanoparticles made of chitosan-g-poly(methyl methacrylate) and poly(vinyl alcohol)-g-poly(methyl methacrylate) and ionotropically crosslinked with sodium tripolyphosphate in the brain after intravenous (i.v.) and i.n. administration to Hsd:ICR mice. After i.v. administration, the highest nanoparticle accumulation was detected in the liver, among other peripheral organs. After i.n. administration of a 10-times smaller nanoparticle dose, the accumulation of the nanoparticles in off-target organs was much lower than after i.v. injection. In particular, the accumulation of the nanoparticles in the liver was 20 times lower than by i.v. When brains were analyzed separately, intravenously administered nanoparticles accumulated mainly in the "top" brain, reaching a maximum after 1 h. Conversely, in i.n. administration, nanoparticles were detected in the "bottom" brain and the head (maximum reached after 2 h) owing to their retention in the nasal mucosa and could serve as a reservoir from which the drug is released and transported to the brain over time. Overall, results indicate that i.n. nanoparticles reach similar brain bioavailability, though with a 10-fold smaller dose, and accumulate in off-target organs to a more limited extent and only after redistribution through the systemic circulation. At the same time, both administration routes seem to lead to differential accumulation in brain regions, and thus, they could be beneficial in the treatment of different medical conditions.
Collapse
|
138
|
Fisher D, Thomas KA, Abdul‐Rasool S. The Synergistic and Neuroprotective Effects of Alcohol–Antioxidant Treatment on Blood–Brain Barrier Endothelial Cells. Alcohol Clin Exp Res 2020; 44:1997-2007. [DOI: 10.1111/acer.14433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 07/23/2020] [Indexed: 12/26/2022]
Affiliation(s)
- David Fisher
- From the Department of Medical Biosciences (DF, KAT, SA‐R) University of the Western Cape Cape Town South Africa
- School of Health Professions (DF) University of Missouri Columbia Missouri
| | - Kelly Angelique Thomas
- From the Department of Medical Biosciences (DF, KAT, SA‐R) University of the Western Cape Cape Town South Africa
| | - Sahar Abdul‐Rasool
- From the Department of Medical Biosciences (DF, KAT, SA‐R) University of the Western Cape Cape Town South Africa
| |
Collapse
|
139
|
Zhao N, Francis NL, Calvelli HR, Moghe PV. Microglia-targeting nanotherapeutics for neurodegenerative diseases. APL Bioeng 2020; 4:030902. [PMID: 32923843 PMCID: PMC7481010 DOI: 10.1063/5.0013178] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
Advances in nanotechnology have enabled the design of nanotherapeutic platforms that could address the challenges of targeted delivery of active therapeutic agents to the central nervous system (CNS). While the majority of previous research studies on CNS nanotherapeutics have focused on neurons and endothelial cells, the predominant resident immune cells of the CNS, microglia, are also emerging as a promising cellular target for neurodegeneration considering their prominent role in neuroinflammation. Under normal physiological conditions, microglia protect neurons by removing pathological agents. However, long-term exposure of microglia to stimulants will cause sustained activation and lead to neuronal damage due to the release of pro-inflammatory agents, resulting in neuroinflammation and neurodegeneration. This Perspective highlights criteria to be considered when designing microglia-targeting nanotherapeutics for the treatment of neurodegenerative disorders. These criteria include conjugating specific microglial receptor-targeting ligands or peptides to the nanoparticle surface to achieve targeted delivery, leveraging microglial phagocytic properties, and utilizing biocompatible and biodegradable nanomaterials with low immune reactivity and neurotoxicity. In addition, certain therapeutic agents for the controlled inhibition of toxic protein aggregation and for modulation of microglial activation pathways can also be incorporated within the nanoparticle structure without compromising stability. Overall, considering the multifaceted disease mechanisms of neurodegeneration, microglia-targeted nanodrugs and nanotherapeutic particles may have the potential to resolve multiple pathological determinants of the disease and to guide a shift in the microglial phenotype spectrum toward a more neuroprotective state.
Collapse
Affiliation(s)
- Nanxia Zhao
- Department of Chemical and Biochemical Engineering, 98 Brett Rd., Rutgers University, Piscataway, New Jersey 08854, USA
| | - Nicola L. Francis
- Department of Biomedical Engineering, 599 Taylor Rd., Rutgers University, Piscataway, New Jersey 08854, USA
| | - Hannah R. Calvelli
- Department of Molecular Biology and Biochemistry, 604 Allison Rd., Rutgers University, Piscataway, New Jersey 08854, USA
| | | |
Collapse
|
140
|
Tu TW, Kovacs ZI, Sundby M, Witko JA, Papadakis GZ, Reid WC, Hammoud DA, Frank JA. Diffusion Tensor Imaging and Chemical Exchange Saturation Transfer MRI Evaluation on the Long-Term Effects of Pulsed Focused Ultrasound and Microbubbles Blood Brain Barrier Opening in the Rat. Front Neurosci 2020; 14:908. [PMID: 32982680 PMCID: PMC7478124 DOI: 10.3389/fnins.2020.00908] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/05/2020] [Indexed: 11/16/2022] Open
Abstract
Blood-brain barrier opening (BBBO) with pulsed Focused Ultrasound (pFUS) and microbubbles (MB) has received increasing interest as a method for neurotherapeutics of the central nervous system. In general, conventional MRI [i.e., T2w, T2∗w, gadolinium (Gd) enhanced T1w] is used to monitor the effects of pFUS+MB on BBBO and/or assess whether sonication results in parenchymal damage. This study employed multimodal MRI techniques and 18F-Fludeoxyglucose (FDG) PET to evaluate the effects of single and multiple weekly pFUS+MB sessions on morphology and glucose utilization levels in the rat cortex and hippocampus. pFUS was performed with 0.548 MHz transducer with a slow infusion over 1 min of OptisonTM (5–8 × 107 MB) in nine focal points in cortex and four in hippocampus. During pFUS+MB treatment, Gd-T1w was performed at 3 T to confirm BBBO, along with subsequent T2w, T2∗w, DTI and glucose CEST (glucoCEST)-weighted imaging by high field 9.4 T and compared with FDG-PET and immunohistochemistry. Animals receiving a single pFUS+MB exhibited minimal hypointense voxels on T2∗w. Brains receiving multiple pFUS+MB treatments demonstrated persistent T2w and T2∗ abnormalities associated with changes in DTI and glucoCEST when compared to contralateral parenchyma. Decreased glucoCEST contrast was substantiated by FDG-PET in cortex following multiple sonications. Immunohistochemistry showed significantly dilated vessels and decreased neuronal glucose transporter (GLUT3) expression in sonicated cortex and hippocampus without changes in neuronal counts. These results suggest the importance to standardize MRI protocols in concert with advanced imaging techniques when evaluating long term effects of pFUS+MB BBBO in clinical trials for neurological diseases.
Collapse
Affiliation(s)
- Tsang-Wei Tu
- Molecular Imaging Laboratory, Department of Radiology, Howard University College of Medicine, Washington, DC, United States.,Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States.,Center for Neuroscience and Regenerative Medicine, Henry Jackson Foundation, Bethesda, MD, United States
| | - Zsofia I Kovacs
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States.,Institute for Biomedical Engineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Maggie Sundby
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Jaclyn A Witko
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States.,Center for Neuroscience and Regenerative Medicine, Henry Jackson Foundation, Bethesda, MD, United States
| | - Georgios Z Papadakis
- Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States
| | - William C Reid
- Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health (NIH), Bethesda, MD, United States.,National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
141
|
Tan J, Sun W, Lu L, Xiao Z, Wei H, Shi W, Wang Y, Han S, Shuai X. I6P7 peptide modified superparamagnetic iron oxide nanoparticles for magnetic resonance imaging detection of low-grade brain gliomas. J Mater Chem B 2020; 7:6139-6147. [PMID: 31553351 DOI: 10.1039/c9tb01563a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Glioma, the most severe primary brain malignancy, has very low survival rates and a high level of recurrence. Nowadays, conventional treatments for these patients are suffering a similar plight owing to the distinctive features of the malignant gliomas, for example chemotherapy is limited by the blood-brain barrier while surgery and radiation therapy are affected by the unclear boundaries of tumor from normal tissue. In the present study, a novel superparamagnetic iron oxide (SPIO) nanoprobe for enhanced T2-weighted magnetic resonance imaging (MRI) was developed. A frequently used MRI probe, SPIO nanoparticles, was coated with a silica outer layer and for the first time was covalently modified with interleukin-6 receptor targeting peptides (I6P7) to promote transportation through the blood-brain barrier and recognition of low-grade gliomas. The efficiency of transcytosis across the blood-brain barrier was examined in vitro using a transwell invasion model and in vivo in nude mice with orthotopic low-grade gliomas. The targeting nanoprobe showed significant MRI enhancement and has potential for use in the diagnosis of low-grade gliomas.
Collapse
Affiliation(s)
- Junyi Tan
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Coutinho MF, Santos JI, S. Mendonça L, Matos L, Prata MJ, S. Jurado A, Pedroso de Lima MC, Alves S. Lysosomal Storage Disease-Associated Neuropathy: Targeting Stable Nucleic Acid Lipid Particle (SNALP)-Formulated siRNAs to the Brain as a Therapeutic Approach. Int J Mol Sci 2020; 21:ijms21165732. [PMID: 32785133 PMCID: PMC7461213 DOI: 10.3390/ijms21165732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
More than two thirds of Lysosomal Storage Diseases (LSDs) present central nervous system involvement. Nevertheless, only one of the currently approved therapies has an impact on neuropathology. Therefore, alternative approaches are under development, either addressing the underlying enzymatic defect or its downstream consequences. Also under study is the possibility to block substrate accumulation upstream, by promoting a decrease of its synthesis. This concept is known as substrate reduction therapy and may be triggered by several molecules, such as small interfering RNAs (siRNAs). siRNAs promote RNA interference, a naturally occurring sequence-specific post-transcriptional gene-silencing mechanism, and may target virtually any gene of interest, inhibiting its expression. Still, naked siRNAs have limited cellular uptake, low biological stability, and unfavorable pharmacokinetics. Thus, their translation into clinics requires proper delivery methods. One promising platform is a special class of liposomes called stable nucleic acid lipid particles (SNALPs), which are characterized by high cargo encapsulation efficiency and may be engineered to promote targeted delivery to specific receptors. Here, we review the concept of SNALPs, presenting a series of examples on their efficacy as siRNA nanodelivery systems. By doing so, we hope to unveil the therapeutic potential of these nanosystems for targeted brain delivery of siRNAs in LSDs.
Collapse
Affiliation(s)
- Maria Francisca Coutinho
- Research and Development Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA I.P), Rua Alexandre Herculano, 321, 4000-055 Porto, Portugal; (J.I.S.); (L.M.); (S.A.)
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- Correspondence: ; Tel.: +351-(223)-401-113
| | - Juliana Inês Santos
- Research and Development Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA I.P), Rua Alexandre Herculano, 321, 4000-055 Porto, Portugal; (J.I.S.); (L.M.); (S.A.)
- Biology Department, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal;
| | - Liliana S. Mendonça
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (L.S.M.); (M.C.P.d.L.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Liliana Matos
- Research and Development Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA I.P), Rua Alexandre Herculano, 321, 4000-055 Porto, Portugal; (J.I.S.); (L.M.); (S.A.)
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
| | - Maria João Prata
- Biology Department, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal;
- i3S—Institute of Research and Innovation in Health/IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal
| | - Amália S. Jurado
- University of Coimbra, CNC—Center for Neuroscience and Cell Biology, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal;
| | - Maria C. Pedroso de Lima
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (L.S.M.); (M.C.P.d.L.)
| | - Sandra Alves
- Research and Development Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA I.P), Rua Alexandre Herculano, 321, 4000-055 Porto, Portugal; (J.I.S.); (L.M.); (S.A.)
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
| |
Collapse
|
143
|
Tan L, Wang Y, Jiang Y, Wang R, Zu J, Tan R. Hydroxysafflor Yellow A Together with Blood-Brain Barrier Regulator Lexiscan for Cerebral Ischemia Reperfusion Injury Treatment. ACS OMEGA 2020; 5:19151-19164. [PMID: 32775917 PMCID: PMC7408215 DOI: 10.1021/acsomega.0c02502] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/10/2020] [Indexed: 05/10/2023]
Abstract
Pharmacodynamic and biodistribution effects are two important factors in drug research. As a clinical drug, the neuroprotective effects and mechanisms of hydroxysafflor yellow A (HSYA) have been widely reported but have still not been described in enough detail. In this study, we first aimed to improve the pharmacology of HSYA in nerve injury treatments. The down-regulative expression of cytokines, including NLRP3, ASC, Caspase-1, GSDMD, IL-1β, IL-18, LDH, NF-κB, and p-p56, suggested that HSYA could both suppress pyroptosis and apoptosis pathway activation during the nerve injury. Additionally, HSYA improved the cellular viability in an oxidative stress damage cell model. Second, to further improve the therapeutic effect of the HSYA, we tried to enhance the concentration of HSYA in a lesion. The FDA-approved adenosine receptor agonist Lexiscan (Lex) could inhibit the expression of P-glycoprotein on the endothelial cell surface to transiently increase the permeability of the blood-brain barrier (BBB) without any sustained damage, which was used to assist HSYA in passing through the BBB to increase the accumulation in the brain. Furthermore, living image and distribution detection in vivo showed that the accumulation of HSYA in the brain could be significantly increased with the addition of Lex. Lastly, HSYA together with Lex (Lex-HSYA) could significantly reduce the volume of cerebral infarction, improve the histopathological morphology, and recruit brain-derived neurotrophic factors to alleviate the cerebral ischemia reperfusion injury. In conclusion, the pyroptosis pathway could act as a novel therapeutic target of HSYA in nerve injury treatment, and Lex-HSYA could be a promising candidate for nerve injury treatments.
Collapse
Affiliation(s)
- Liwei Tan
- College
of Life Science and Engineering, Southwest
Jiaotong University, Chengdu 610031, China
| | - Yeye Wang
- College
of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Yu Jiang
- College
of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Rong Wang
- College
of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Jingzhi Zu
- College
of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Rui Tan
- College
of Life Science and Engineering, Southwest
Jiaotong University, Chengdu 610031, China
- or . Phone/Fax: +86-28-87634667
| |
Collapse
|
144
|
Modification of curcumin-loaded liposome with edible compounds to enhance ability of crossing blood brain barrier. Colloids Surf A Physicochem Eng Asp 2020. [DOI: 10.1016/j.colsurfa.2020.124862] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
145
|
Lewicky JD, Fraleigh NL, Boraman A, Martel AL, Nguyen TMD, Schiller PW, Shiao TC, Roy R, Montaut S, Le HT. Mannosylated glycoliposomes for the delivery of a peptide kappa opioid receptor antagonist to the brain. Eur J Pharm Biopharm 2020; 154:290-296. [PMID: 32717389 DOI: 10.1016/j.ejpb.2020.07.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 06/03/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
Abstract
Dynantin is a potent and selective synthetic polypeptide kappa opioid receptor antagonist which has potential antidepressant and anxiolytic-like therapeutic applications, however its clinical development has been hampered by plasma stability issues and poor penetration of the blood brain barrier. Targeted liposome delivery systems represent a promising and non-invasive approach to improving the delivery of therapeutic agents across the blood brain barrier. As part of our work focused on targeted drug delivery, we have developed a novel mannosylated liposome system. Herein, we investigate these glycoliposomes for the targeted delivery of dynantin to the central nervous system. Cholesterol was tested and optimized as a formulation excipient, where it improved particle stability as measured via particle size, entrapment and ex vivo plasma stability of dynantin. The in vitro PRESTO-TANGO assay system was used to confirm that glycoliposomal entrapment did not impact the affinity or activity of the peptide at its receptor. Finally, in vivo distribution studies in mice showed that the mannosylated glycoliposomes significantly improved delivery of dynantin to the brain. Overall, the results clearly demonstrate the potential of our glycoliposomes as a targeted delivery system for therapeutic agents of the central nervous system.
Collapse
Affiliation(s)
- Jordan D Lewicky
- Health Sciences North Research Institute, 56 Walford Road, Sudbury, Ontario P3E 2H2, Canada
| | - Nya L Fraleigh
- Health Sciences North Research Institute, 56 Walford Road, Sudbury, Ontario P3E 2H2, Canada
| | - Amanda Boraman
- Health Sciences North Research Institute, 56 Walford Road, Sudbury, Ontario P3E 2H2, Canada; Department of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, Ontario P3E 2C6, Canada
| | - Alexandrine L Martel
- Health Sciences North Research Institute, 56 Walford Road, Sudbury, Ontario P3E 2H2, Canada
| | - Thi M-D Nguyen
- Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montreal, 110 Pine Avenue W, Montreal, Quebec H2W 1R7, Canada
| | - Peter W Schiller
- Laboratory of Chemical Biology and Peptide Research, Clinical Research Institute of Montreal, 110 Pine Avenue W, Montreal, Quebec H2W 1R7, Canada; Department of Pharmacology and Physiology, University of Montreal, 2900 Boulevard Édouard-Montpetit, Montreal, Quebec H3T 1J4, Canada
| | - Tze Chieh Shiao
- Department of Chemistry, Université du Québec à Montréal, Montreal, Quebec H3C 3P8, Canada
| | - René Roy
- Department of Chemistry, Université du Québec à Montréal, Montreal, Quebec H3C 3P8, Canada
| | - Sabine Montaut
- Department of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, Ontario P3E 2C6, Canada; Biomolecular Sciences Programme, Laurentian University, Subdury, Ontario, Canada
| | - Hoang-Thanh Le
- Health Sciences North Research Institute, 56 Walford Road, Sudbury, Ontario P3E 2H2, Canada; Department of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, Ontario P3E 2C6, Canada; Northern Ontario School of Medicine, Medicinal Sciences Division, 935 Ramsey Lake Road, Sudbury, Ontario P3E 2C6, Canada; Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, Ontario P3E 2C6, Canada.
| |
Collapse
|
146
|
Development of an LDL Receptor-Targeted Peptide Susceptible to Facilitate the Brain Access of Diagnostic or Therapeutic Agents. BIOLOGY 2020; 9:biology9070161. [PMID: 32664518 PMCID: PMC7407834 DOI: 10.3390/biology9070161] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/30/2020] [Accepted: 07/08/2020] [Indexed: 12/02/2022]
Abstract
Blood-brain barrier (BBB) crossing and brain penetration are really challenging for the delivery of therapeutic agents and imaging probes. The development of new crossing strategies is needed, and a wide range of approaches (invasive or not) have been proposed so far. The receptor-mediated transcytosis is an attractive mechanism, allowing the non-invasive penetration of the BBB. Among available targets, the low-density lipoprotein (LDL) receptor (LDLR) shows favorable characteristics mainly because of the lysosome-bypassed pathway of LDL delivery to the brain, allowing an intact discharge of the carried ligand to the brain targets. The phage display technology was employed to identify a dodecapeptide targeted to the extracellular domain of LDLR (ED-LDLR). This peptide was able to bind the ED-LDLR in the presence of natural ligands and dissociated at acidic pH and in the absence of calcium, in a similar manner as the LDL. In vitro, our peptide was endocytosed by endothelial cells through the caveolae-dependent pathway, proper to the LDLR route in BBB, suggesting the prevention of its lysosomal degradation. The in vivo studies performed by magnetic resonance imaging and fluorescent lifetime imaging suggested the brain penetration of this ED-LDLR-targeted peptide.
Collapse
|
147
|
Neuroprotection by curcumin: A review on brain delivery strategies. Int J Pharm 2020; 585:119476. [DOI: 10.1016/j.ijpharm.2020.119476] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/05/2020] [Accepted: 05/24/2020] [Indexed: 12/26/2022]
|
148
|
Liu W, Lin Q, Fu Y, Huang S, Guo C, Li L, Wang L, Zhang Z, Zhang L. Target delivering paclitaxel by ferritin heavy chain nanocages for glioma treatment. J Control Release 2020; 323:191-202. [DOI: 10.1016/j.jconrel.2019.12.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/26/2019] [Accepted: 12/09/2019] [Indexed: 01/01/2023]
|
149
|
Ding S, Khan AI, Cai X, Song Y, Lyu Z, Du D, Dutta P, Lin Y. Overcoming blood-brain barrier transport: Advances in nanoparticle-based drug delivery strategies. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2020; 37:112-125. [PMID: 33093794 PMCID: PMC7575138 DOI: 10.1016/j.mattod.2020.02.001] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The Blood-Brain Barrier (BBB), a unique structure in the central nervous system (CNS), protects the brain from bloodborne pathogens by its excellent barrier properties. Nevertheless, this barrier limits therapeutic efficacy and becomes one of the biggest challenges in new drug development for neurodegenerative disease and brain cancer. Recent breakthroughs in nanotechnology have resulted in various nanoparticles (NPs) as drug carriers to cross the BBB by different methods. This review presents the current understanding of advanced NP-mediated non-invasive drug delivery for the treatment of neurological disorders. Herein, the complex compositions and special characteristics of BBB are elucidated exhaustively. Moreover, versatile drug nanocarriers with their recent applications and their pathways on different drug delivery strategies to overcome the formidable BBB obstacle are briefly discussed. In terms of significance, this paper provides a general understanding of how various properties of nanoparticles aid in drug delivery through BBB and usher the development of novel nanotechnology-based nanomaterials for cerebral disease therapies.
Collapse
Affiliation(s)
| | | | - Xiaoli Cai
- School of Mechanical and Materials Engineering, Washington State University, PO Box 642920 Pullman, Washington 99164, United States
| | - Yang Song
- School of Mechanical and Materials Engineering, Washington State University, PO Box 642920 Pullman, Washington 99164, United States
| | - Zhaoyuan Lyu
- School of Mechanical and Materials Engineering, Washington State University, PO Box 642920 Pullman, Washington 99164, United States
| | - Dan Du
- School of Mechanical and Materials Engineering, Washington State University, PO Box 642920 Pullman, Washington 99164, United States
| | - Prashanta Dutta
- School of Mechanical and Materials Engineering, Washington State University, PO Box 642920 Pullman, Washington 99164, United States
| | - Yuehe Lin
- School of Mechanical and Materials Engineering, Washington State University, PO Box 642920 Pullman, Washington 99164, United States
| |
Collapse
|
150
|
Zipfel P, Rochais C, Baranger K, Rivera S, Dallemagne P. Matrix Metalloproteinases as New Targets in Alzheimer's Disease: Opportunities and Challenges. J Med Chem 2020; 63:10705-10725. [PMID: 32459966 DOI: 10.1021/acs.jmedchem.0c00352] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although matrix metalloproteinases (MMPs) are implicated in the regulation of numerous physiological processes, evidence of their pathological roles have also been obtained in the last decades, making MMPs attractive therapeutic targets for several diseases. Recent discoveries of their involvement in central nervous system (CNS) disorders, and in particular in Alzheimer's disease (AD), have paved the way to consider MMP modulators as promising therapeutic strategies. Over the past few decades, diverse approaches have been undertaken in the design of therapeutic agents targeting MMPs for various purposes, leading, more recently, to encouraging developments. In this article, we will present recent examples of inhibitors ranging from small molecules and peptidomimetics to biologics. We will also discuss the scientific knowledge that has led to the development of emerging tools and techniques to overcome the challenges of selective MMP inhibition.
Collapse
Affiliation(s)
- Pauline Zipfel
- Normandie Univ, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Christophe Rochais
- Normandie Univ, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Kévin Baranger
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Santiago Rivera
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Patrick Dallemagne
- Normandie Univ, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| |
Collapse
|