1
|
Sun B, Li R, Ji N, Liu H, Wang H, Chen C, Bai L, Su J, Chen J. Brain-targeting drug delivery systems: The state of the art in treatment of glioblastoma. Mater Today Bio 2025; 30:101443. [PMID: 39866779 PMCID: PMC11759563 DOI: 10.1016/j.mtbio.2025.101443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Glioblastoma (GBM) is the most prevalent primary malignant brain tumor, characterized by a high mortality rate and a poor prognosis. The blood-brain barrier (BBB) and the blood-tumor barrier (BTB) present significant obstacles to the efficacy of tumor-targeted pharmacotherapy, thereby impeding the therapeutic potential of numerous candidate drugs. Targeting delivery of adequate doses of drug across the BBB to treat GBM has become a prominent research area in recent years. This emphasis has driven the exploration and evaluation of diverse technologies for GBM pharmacotherapy, with some already undergoing clinical trials. This review provides a thorough overview of recent advancements and challenges in targeted drug delivery for GBM treatment. It specifically emphasizes systemic drug administration strategies to assess their potential and limitations in GBM treatment. Furthermore, this review highlights promising future research directions in the development of intelligent drug delivery systems aimed at overcoming current challenges and enhancing therapeutic efficacy against GBM. These advancements not only support foundational research on targeted drug delivery systems for GBM but also offer methodological approaches for future clinical applications.
Collapse
Affiliation(s)
- Bo Sun
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Rong Li
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ning Ji
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hongxiang Wang
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chao Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
2
|
Lee Y, Kwon J, Jang M, Ma S, Jun KY, Yoon M, Ye S. Glioblastoma in a paper industry worker exposed to high concentrations of formaldehyde: a case report. Ann Occup Environ Med 2024; 36:e17. [PMID: 39144150 PMCID: PMC11322565 DOI: 10.35371/aoem.2024.36.e17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 08/16/2024] Open
Abstract
Background Formaldehyde was classified as a Group I Carcinogen by the International Agency for Research on Cancer (IARC) in 2006. While the IARC has stated that there is a lack of evidence that formaldehyde causes brain cancer, three meta-analyses have consistently reported a significantly higher risk of brain cancer in workers exposed to high levels of formaldehyde. Therefore, we report a case of a worker who was diagnosed with glioblastoma after being exposed to high concentrations of formaldehyde while working with formaldehyde resin in the paper industry. Case presentation A 40-year-old male patient joined an impregnated paper manufacturer and performed impregnation work using formaldehyde resin for 10 years and 2 months. In 2017, the patient experienced a severe headache and visited the hospital for brain magnetic resonance imaging, which revealed a mass. In the same year, the patient underwent a craniotomy for brain tumor resection and was diagnosed with glioblastoma of the temporal lobe. In 2019, a craniotomy was performed owing to the recurrence of the brain tumor, but he died in 2020. An exposure assessment of the work environment determined that the patient was exposed to formaldehyde above the exposure threshold of 0.3 ppm continuously for more than 10 years and that he had high respiratory and dermal exposure through performing work without wearing a respirator or protective gloves. Conclusions This case report represents the first instance where the epidemiological investigation and evaluation committee of the Occupational Safety and Health Research Institute in Korea recognized the scientific evidence of work-related brain tumors due to long-term exposure to high concentrations of formaldehyde during impregnated paperwork. This case highlights the importance of proper workplace management, informing workers that prolonged exposure to formaldehyde in impregnation work can cause brain tumors and minimizing exposure in similar processes.
Collapse
Affiliation(s)
- Youngshin Lee
- Occupational Safety and Health Research Institute, Korea Occupational Safety and Health Agency, Ulsan, Korea
| | - Jiwoon Kwon
- Occupational Safety and Health Research Institute, Korea Occupational Safety and Health Agency, Incheon, Korea
- National Fire Research Institute, Asan, Korea
| | - Miyeon Jang
- Occupational Safety and Health Research Institute, Korea Occupational Safety and Health Agency, Ulsan, Korea
| | - Seongwon Ma
- Occupational Safety and Health Research Institute, Korea Occupational Safety and Health Agency, Ulsan, Korea
- Department of Occupational and Environmental Medicine, Osan Hankook Hospital, Osan, Korea
| | - Kyo Yeon Jun
- Occupational Safety and Health Research Institute, Korea Occupational Safety and Health Agency, Incheon, Korea
| | - Minjoo Yoon
- Occupational Safety and Health Research Institute, Korea Occupational Safety and Health Agency, Incheon, Korea
| | - Shinhee Ye
- Occupational Safety and Health Research Institute, Korea Occupational Safety and Health Agency, Incheon, Korea
| |
Collapse
|
3
|
Tirgar F, Azizi Z, Hadjighassem M. A Novel Approach for Mucosal and Bulbar Olfactory Ensheathing Cells Isolation Based on the Non-adherent Subculture Technique. Basic Clin Neurosci 2024; 15:211-220. [PMID: 39228451 PMCID: PMC11367208 DOI: 10.32598/bcn.2022.3579.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/15/2022] [Accepted: 04/12/2022] [Indexed: 09/05/2024] Open
Abstract
Introduction Olfactory ensheathing cells (OECs) are widely used in transplantation studies. The high purification of this unique cell type is valuable for medical applications. Although recent improvements in OECs isolation procedures opened a new era in this field, the high purification efficacy and viability rate are still of concern. The most widely used OECs isolation techniques can be broadly classified based on adherence properties, particularly in olfactory bulb-derived OEC isolation. Considering the invasive nature of harvesting OECs from human olfactory bulbs, a highly efficient purification of these cells from olfactory mucosa can benefit clinical trials. In this study, we isolated OECs from rats' olfactory bulbs and mucosa due to their differential adherence properties and compared them. Methods Cell preparations were characterized by NGFR p75 and S100β antibodies, the specific markers for OECs, using immunocytochemistry and western blot analysis, respectively. OECs morphology and viability were monitored over time by microscopy and MTT (3-[4,5-dimethylthiazol2-yl]-2,5-diphenyltetrazolium bromide) assay. Results We found that OECs could be purified from the olfactory mucosa using our suggested method as efficiently as the olfactory bulb. Both derived OECs showed high levels of NGFR p75 and S100β expression, although the S100β expression was higher in olfactory mucosa-derived OECs preparations (P<0.05). Moreover, there was no significant difference between the two sources in cell viability in our suggested protocol. Conclusion Due to the non-invasive harvesting method, olfactory mucosa-derived OECs are preferred from a clinical point of view in transplantation studies.
Collapse
Affiliation(s)
- Fatemeh Tirgar
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Azizi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Hosseindoost S, Dehpour AR, Dehghan S, Javadi SAH, Arjmand B, Fallah A, Hadjighassem M. Fluoxetine enhances the antitumor effect of olfactory ensheathing cell-thymidine kinase/ganciclovir gene therapy in human glioblastoma multiforme cells through upregulation of Connexin43 levels. Drug Dev Res 2023; 84:1739-1750. [PMID: 37769152 DOI: 10.1002/ddr.22119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/30/2023]
Abstract
Glioblastoma multiforme (GBM) is the most invasive form of primary brain astrocytoma, resulting in poor clinical outcomes. Herpes simplex virus thymidine kinase/ganciclovir (HSV-TK/GCV) gene therapy is considered a promising strategy for GBM treatment. Since Connexin43 (Cx43) expression is reduced in GBM cells, increasing Cx43 levels could enhance the effectiveness of gene therapy. The present study aims to examine the impact of fluoxetine on HSV-TK/GCV gene therapy in human GBM cells using human olfactory ensheathing cells (OECs) as vectors. The effect of fluoxetine on Cx43 levels was assessed using the western blot technique. GBM-derived astrocytes and OECs-TK were Cocultured, and the effect of fluoxetine on the Antitumor effect of OEC-TK/GCV gene therapy was evaluated using MTT assay and flow cytometry. Our results showed that fluoxetine increased Cx43 levels in OECs and GBM cells and augmented the killing effect of OECs-TK on GBM cells. Western blot data revealed that fluoxetine enhanced the Bax/Bcl2 ratio and the levels of cleaved caspase-3 in the coculture of OECs-TK and GBM cells. Moreover, flow cytometry data indicated that fluoxetine increased the percentage of apoptotic cells in the coculture system. This study suggests that fluoxetine, by upregulating Cx43 levels, could strengthen the Antitumor effect of OEC-TK/GCV gene therapy on GBM cells.
Collapse
Affiliation(s)
- Saereh Hosseindoost
- Pain Research Center, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Cord Injury Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad R Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Dehghan
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed A H Javadi
- Brain and Spinal Cord Injury Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Neurosurgery Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Fallah
- Space Medicine B.V., Rotterdam, the Netherlands
- Systems and Synthetic Biology Group, Mede Bioeconomy Company, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Faisal SM, Castro MG, Lowenstein PR. Combined cytotoxic and immune-stimulatory gene therapy using Ad-TK and Ad-Flt3L: Translational developments from rodents to glioma patients. Mol Ther 2023; 31:2839-2860. [PMID: 37574780 PMCID: PMC10556227 DOI: 10.1016/j.ymthe.2023.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/14/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023] Open
Abstract
Gliomas are the most prevalent and devastating primary malignant brain tumors in adults. Despite substantial advances in understanding glioma biology, there have been no regulatory drug approvals in the US since bevacizumab in 2009 and tumor treating fields in 2011. Recent phase III clinical trials have failed to meet their prespecified therapeutic primary endpoints, highlighting the need for novel therapies. The poor prognosis of glioma patients, resistance to chemo-radiotherapy, and the immunosuppressive tumor microenvironment underscore the need for the development of novel therapies. Gene therapy-based immunotherapeutic strategies that couple the ability of the host immune system to specifically kill glioma cells and develop immunological memory have shown remarkable progress. Two adenoviral vectors expressing Ad-HSV1-TK/GCV and Ad-Flt3L have shown promising preclinical data, leading to FDA approval of a non-randomized, phase I open-label, first in human trial to test safety, cytotoxicity, and immune-stimulatory efficiency in high-grade glioma patients (NCT01811992). This review provides a thorough overview of immune-stimulatory gene therapy highlighting recent advancements, potential drawbacks, future directions, and recommendations for future implementation of clinical trials.
Collapse
Affiliation(s)
- Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Rogel Cancer Centre, University of Michigan Medical School, Ann Arbor, MI 48108, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Rogel Cancer Centre, University of Michigan Medical School, Ann Arbor, MI 48108, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Rogel Cancer Centre, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48108, USA.
| |
Collapse
|
6
|
Boyuklieva R, Zagorchev P, Pilicheva B. Computational, In Vitro, and In Vivo Models for Nose-to-Brain Drug Delivery Studies. Biomedicines 2023; 11:2198. [PMID: 37626694 PMCID: PMC10452071 DOI: 10.3390/biomedicines11082198] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Direct nose-to-brain drug delivery offers the opportunity to treat central nervous system disorders more effectively due to the possibility of drug molecules reaching the brain without passing through the blood-brain barrier. Such a delivery route allows the desired anatomic site to be reached while ensuring drug effectiveness, minimizing side effects, and limiting drug losses and degradation. However, the absorption of intranasally administered entities is a complex process that considerably depends on the interplay between the characteristics of the drug delivery systems and the nasal mucosa. Various preclinical models (in silico, in vitro, ex vivo, and in vivo) are used to study the transport of drugs after intranasal administration. The present review article attempts to summarize the different computational and experimental models used so far to investigate the direct delivery of therapeutic agents or colloidal carriers from the nasal cavity to the brain tissue. Moreover, it provides a critical evaluation of the data available from different studies and identifies the advantages and disadvantages of each model.
Collapse
Affiliation(s)
- Radka Boyuklieva
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Plamen Zagorchev
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Bissera Pilicheva
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| |
Collapse
|
7
|
Rufino-Ramos D, Albuquerque PR, Leandro K, Carmona V, Martins IM, Fernandes R, Henriques C, Lobo D, Faro R, Perfeito R, Mendonça LS, Pereira D, Gomes CM, Nobre RJ, Pereira de Almeida L. Extracellular vesicle-based delivery of silencing sequences for the treatment of Machado-Joseph disease/spinocerebellar ataxia type 3. Mol Ther 2023; 31:1275-1292. [PMID: 37025062 PMCID: PMC10188911 DOI: 10.1016/j.ymthe.2023.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Machado-Joseph disease (MJD)/spinocerebellar ataxia type 3 (SCA3) is the most common autosomal dominantly inherited ataxia worldwide. It is caused by an over-repetition of the trinucleotide CAG within the ATXN3 gene, which confers toxic properties to ataxin-3 (ATXN3) species. RNA interference technology has shown promising therapeutic outcomes but still lacks a non-invasive delivery method to the brain. Extracellular vesicles (EVs) emerged as promising delivery vehicles due to their capacity to deliver small nucleic acids, such as microRNAs (miRNAs). miRNAs were found to be enriched into EVs due to specific signal motifs designated as ExoMotifs. In this study, we aimed at investigating whether ExoMotifs would promote the packaging of artificial miRNAs into EVs to be used as non-invasive therapeutic delivery vehicles to treat MJD/SCA3. We found that miRNA-based silencing sequences, associated with ExoMotif GGAG and ribonucleoprotein A2B1 (hnRNPA2B1), retained the capacity to silence mutant ATXN3 (mutATXN3) and were 3-fold enriched into EVs. Bioengineered EVs containing the neuronal targeting peptide RVG on the surface significantly decreased mutATXN3 mRNA in primary cerebellar neurons from MJD YAC 84.2 and in a novel dual-luciferase MJD mouse model upon daily intranasal administration. Altogether, these findings indicate that bioengineered EVs carrying miRNA-based silencing sequences are a promising delivery vehicle for brain therapy.
Collapse
Affiliation(s)
- David Rufino-Ramos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Patrícia R Albuquerque
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Kevin Leandro
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Vitor Carmona
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Inês M Martins
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Rita Fernandes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Carina Henriques
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Diana Lobo
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Rosário Faro
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Rita Perfeito
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Liliana S Mendonça
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Dina Pereira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Célia M Gomes
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Rui Jorge Nobre
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
8
|
Paes-Colli Y, Trindade PMP, Vitorino LC, Piscitelli F, Iannotti FA, Campos RMP, Isaac AR, de Aguiar AFL, Allodi S, de Mello FG, Einicker-Lamas M, de Siqueira-Santos R, Di Marzo V, Tannous BA, Carvalho LA, De Melo Reis RA, Sampaio LS. Activation of cannabinoid type 1 receptor (CB1) modulates oligodendroglial process branching complexity in rat hippocampal cultures stimulated by olfactory ensheathing glia-conditioned medium. Front Cell Neurosci 2023; 17:1134130. [PMID: 37138770 PMCID: PMC10150319 DOI: 10.3389/fncel.2023.1134130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/16/2023] [Indexed: 05/05/2023] Open
Abstract
The endocannabinoid system (ECS) refers to a complex cell-signaling system highly conserved among species formed by numerous receptors, lipid mediators (endocannabinoids) and synthetic and degradative enzymes. It is widely distributed throughout the body including the CNS, where it participates in synaptic signaling, plasticity and neurodevelopment. Besides, the olfactory ensheathing glia (OEG) present in the olfactory system is also known to play an important role in the promotion of axonal growth and/or myelination. Therefore, both OEG and the ECS promote neurogenesis and oligodendrogenesis in the CNS. Here, we investigated if the ECS is expressed in cultured OEG, by assessing the main markers of the ECS through immunofluorescence, western blotting and qRT-PCR and quantifying the content of endocannabinoids in the conditioned medium of these cells. After that, we investigated whether the production and release of endocannabinoids regulate the differentiation of oligodendrocytes co-cultured with hippocampal neurons, through Sholl analysis in oligodendrocytes expressing O4 and MBP markers. Additionally, we evaluated through western blotting the modulation of downstream pathways such as PI3K/Akt/mTOR and ERK/MAPK, being known to be involved in the proliferation and differentiation of oligodendrocytes and activated by CB1, which is the major endocannabinoid responsive receptor in the brain. Our data show that OEG expresses key genes of the ECS, including the CB1 receptor, FAAH and MAGL. Besides, we were able to identify AEA, 2-AG and AEA related mediators palmitoylethanolamide (PEA) and oleoylethanolamide (OEA), in the conditioned medium of OEG cultures. These cultures were also treated with URB597 10-9 M, a FAAH selective inhibitor, or JZL184 10-9 M, a MAGL selective inhibitor, which led to the increase in the concentrations of OEA and 2-AG in the conditioned medium. Moreover, we found that the addition of OEG conditioned medium (OEGCM) enhanced the complexity of oligodendrocyte process branching in hippocampal mixed cell cultures and that this effect was inhibited by AM251 10-6 M, a CB1 receptor antagonist. However, treatment with the conditioned medium enriched with OEA or 2-AG did not alter the process branching complexity of premyelinating oligodendrocytes, while decreased the branching complexity in mature oligodendrocytes. We also observed no change in the phosphorylation of Akt and ERK 44/42 in any of the conditions used. In conclusion, our data show that the ECS modulates the number and maturation of oligodendrocytes in hippocampal mixed cell cultures.
Collapse
Affiliation(s)
- Yolanda Paes-Colli
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila M. P. Trindade
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Louise C. Vitorino
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli, Italy
| | - Fabio Arturo Iannotti
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli, Italy
| | - Raquel M. P. Campos
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alinny R. Isaac
- Laboratório de Doenças Neurodegenerativas, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrey Fabiano Lourenço de Aguiar
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Silvana Allodi
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando G. de Mello
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Einicker-Lamas
- Laboratório de Biomembranas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raphael de Siqueira-Santos
- Laboratório de Agregação de Proteínas e Amiloidoses, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli, Italy
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis, Laval University, Quebec, QC, Canada
| | - Bakhos A. Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Massachusetts General Hospital, Boston, MA, United States
- Neuroscience Program, Harvard Medical School, Boston, MA, United States
| | - Litia A. Carvalho
- Experimental Therapeutics and Molecular Imaging Laboratory, Massachusetts General Hospital, Boston, MA, United States
- Neuroscience Program, Harvard Medical School, Boston, MA, United States
| | - Ricardo A. De Melo Reis
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luzia S. Sampaio
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Luzia S. Sampaio,
| |
Collapse
|
9
|
Designing a Clinical Trial with Olfactory Ensheathing Cell Transplantation-Based Therapy for Spinal Cord Injury: A Position Paper. Biomedicines 2022; 10:biomedicines10123153. [PMID: 36551909 PMCID: PMC9776288 DOI: 10.3390/biomedicines10123153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) represents an urgent unmet need for clinical reparative therapy due to its largely irreversible and devastating effects on patients, and the tremendous socioeconomic burden to the community. While different approaches are being explored, therapy to restore the lost function remains unavailable. Olfactory ensheathing cell (OEC) transplantation is a promising approach in terms of feasibility, safety, and limited efficacy; however, high variability in reported clinical outcomes prevent its translation despite several clinical trials. The aims of this position paper are to present an in-depth analysis of previous OEC transplantation-based clinical trials, identify existing challenges and gaps, and finally propose strategies to improve standardization of OEC therapies. We have reviewed the study design and protocols of clinical trials using OEC transplantation for SCI repair to investigate how and why the outcomes show variability. With this knowledge and our experience as a team of biologists and clinicians with active experience in the field of OEC research, we provide recommendations regarding cell source, cell purity and characterisation, transplantation dosage and format, and rehabilitation. Ultimately, this position paper is intended to serve as a roadmap to design an effective clinical trial with OEC transplantation-based therapy for SCI repair.
Collapse
|
10
|
Tirgar F, Azizi Z, Hosseindoost S, Hadjighassem M. Preclinical gene therapy in glioblastoma multiforme: Using olfactory ensheathing cells containing a suicide gene. Life Sci 2022; 311:121132. [DOI: 10.1016/j.lfs.2022.121132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
|
11
|
Tu YK, Hsueh YH, Huang HC. Human olfactory ensheathing cell-derived extracellular vesicles: miRNA profile and neuroprotective effect. Curr Neurovasc Res 2021; 18:395-408. [PMID: 34645375 DOI: 10.2174/1567202618666211012162111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Extracellular vesicle (EV)-based therapy has been identified as a leading alternative approach in several disease models. EV derived from the olfactory ensheathing cell (OEC) has been documented for its strong neuro-regenerative capacity. However, no information on its cargo that may contribute to its therapeutic effect has been available. OBJECTIVE To report the first miRNA profile of human OEC (hOEC) -EV, and investigate the neuroprotective effects. METHODS hOEC-EV was isolated and sequenced. We established in vitro experiments to assess the therapeutic potential of hOEC-EVs with respect to insulted neural progenitor cells (NPCs), and the angiogenesis effect. Secondary post-injury insults were imitated using t-BHP-mediated oxidative stress. RESULTS We noted a strong abundance of hOEC-EV-miRNAs, including hsa-miR148a-3p, has-miR151a-3p and several members of let-7 family. The common targets of 15 miRNAs among the top 20 miRNAs were thrombospondin 1 and cyclin dependent kinase 6. We demonstrated that hOEC-EVs promote normal NPC proliferation and differentiation to neuron-like morphologies with prolonged axons. hOEC-EVs protect cells from t-BHP mediated apoptosis. We also found that the migration rate of either NPCs or endothelial cells significantly improved with hOEC-EVs. Furthermore, in vitro tube formation assays indicated that angiogenesis, an important process for tissue repair, was significantly enhanced in human umbilical vein endothelial cells exposed to hOEC-EVs. CONCLUSION Our results revealed that hOEC-EVs exert neuroprotective effects by protecting cells from apoptosis and promoting in vitro biological processes that are important to neural tissue repair, including neural cell proliferation, axonal growth, and cell migration, in addition to enhancing angiogenesis. </p>.
Collapse
Affiliation(s)
- Yuan-Kun Tu
- Department of Orthopedic Surgery, E-Da Hospitall, I-Shou University, Kaohsiung city. Taiwan
| | - Yu-Huan Hsueh
- Department of Orthopedic Surgery, E-Da Hospitall, I-Shou University, Kaohsiung city. Taiwan
| | - Hsien-Chang Huang
- Department of Orthopedic Surgery, E-Da Hospitall, I-Shou University, Kaohsiung city. Taiwan
| |
Collapse
|
12
|
Tang T, Chang B, Zhang M, Sun T. Nanoprobe-mediated precise imaging and therapy of glioma. NANOSCALE HORIZONS 2021; 6:634-650. [PMID: 34110340 DOI: 10.1039/d1nh00182e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Gliomas are the most common primary brain tumors in adults, accounting for 80% of primary intracranial tumors. Due to the heterogeneous and infiltrating nature of malignant gliomas and the hindrance of the blood-brain barrier (BBB), it is very difficult to accurately image and differentiate the malignancy grade of gliomas, thus significantly influencing the diagnostic accuracy and subsequent surgery or therapy. In recent years, the rapid development of emerging nanoprobes has provided a promising opportunity for the diagnosis and treatment of gliomas. After rational component regulation and surface modification, functional nanoprobes could efficiently cross the BBB, target gliomas, and realize single-modal or multimodal imaging of gliomas with high clarity. Moreover, these contrast nanoagents could also be conjugated with therapeutic drugs and cure cancerous tissues at the same time. Herein, we focus on the design strategies of nanoprobes for effective crossing of the BBB, and introduce the recent advances in the precise imaging and therapy of gliomas using functional nanoprobes. Finally, we also discuss the challenges and future directions of nanoprobe-based diagnosis and treatment of gliomas.
Collapse
Affiliation(s)
- Tao Tang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China.
| | - Baisong Chang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China.
| | - Mingxi Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China.
| | - Taolei Sun
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China. and School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, P. R. China
| |
Collapse
|
13
|
Rana I, Rieswijk L, Steinmaus C, Zhang L. Formaldehyde and Brain Disorders: A Meta-Analysis and Bioinformatics Approach. Neurotox Res 2021; 39:924-948. [PMID: 33400181 PMCID: PMC8102312 DOI: 10.1007/s12640-020-00320-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
While there is significant investigation and investment in brain and neurodegenerative disease research, current understanding of the etiologies of illnesses like Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and brain cancer remains limited. Environmental exposure to the pollutant formaldehyde, an emerging neurotoxin widely used in industry, is suspected to play a critical role in mediating these disorders, although findings are limited and inconsistent. Focusing on highly exposed groups, we performed a meta-analysis of human epidemiological studies of formaldehyde and neurodegenerative disease (N = 19) or brain tumors (N = 12). To assess the biological plausibility of observed associations, we then conducted a bioinformatics analysis using WikiPathways and the Comparative Toxicogenomics Database and identified candidate genes and pathways that may be related to these interactions. We reported the meta-relative risk (meta-RR) of ALS following high exposures to formaldehyde was increased by 78% (meta-RR = 1.78, 95% confidence interval, CI 1.20-2.65). Similarly, the meta-RR for brain cancer was increased by 71% (meta-RR = 1.71; 95% CI 1.07-2.73) among highly exposed individuals. Multiple sensitivity analyses did not reveal sources of heterogeneity or bias. Our bioinformatics analysis revealed that the oxidative stress genes superoxide dismutase (SOD1, SOD2) and the pro-inflammatory marker tumor necrosis factor (TNF) were identified as the top relevant genes, and the folate metabolism, vitamin B12 metabolism, and the ALS pathways were highly affected by formaldehyde and related to the most brain diseases of interest. Further inquiry revealed the two metabolic pathways are also intimately tied with the formaldehyde cycle. Overall, our bioinformatics analysis supports the link of formaldehyde exposure to ALS or brain tumor reported from our meta-analysis. This new multifactorial approach enabled us to both interrogate the robustness of the epidemiological data and identify genes and pathways that may be involved in these interactions, ultimately lending strong evidence and potential biological plausibility for the association between formaldehyde exposure and brain disease.
Collapse
Affiliation(s)
- Iemaan Rana
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Linda Rieswijk
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
- Institute of Data Science, Maastricht University, Maastricht, Netherlands
| | - Craig Steinmaus
- Division of Epidemiology and Biostatistics, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA.
| |
Collapse
|
14
|
Teng J, Lashgari G, Tabet EI, Tannous BA. The natural compound obtusaquinone targets pediatric high-grade gliomas through ROS-mediated ER stress. Neurooncol Adv 2020; 2:vdaa106. [PMID: 33134921 PMCID: PMC7592425 DOI: 10.1093/noajnl/vdaa106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Pediatric high-grade gliomas (pHGGs) are aggressive primary brain tumors with local invasive growth and poor clinical prognosis. Treatment of pHGGs is particularly challenging given the intrinsic resistance to chemotherapy, an absence of novel therapeutics, and the difficulty of drugs to reach the tumor beds. Accumulating evidence suggests that production of reactive oxygen species (ROS) and misfolded proteins, which typically leads to endoplasmic reticulum (ER) stress, is an essential mechanism in cancer cell survival. Methods Several cell viability assays were used in 6 patient-derived pHGG cultures to evaluate the effect of the natural compound obtusaquinone (OBT) on cytotoxicity. Orthotopic mouse models were used to determine OBT effects in vivo. Immunoblotting, immunostaining, flow cytometry, and biochemical assays were used to investigate the OBT mechanism of action. Results OBT significantly inhibited cell survival of patient-derived pHGG cells in culture. OBT inhibited tumor growth and extended survival in 2 different orthotopic xenograft models. Mechanistically, OBT induced ER stress through abnormal ROS accumulation. Conclusion Our data demonstrate the utility and feasibility of OBT as a potential therapeutic option for improving the clinical treatment of pHGGs.
Collapse
Affiliation(s)
- Jian Teng
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Ghazal Lashgari
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Elie I Tabet
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Unit, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Veronesi MC, Alhamami M, Miedema SB, Yun Y, Ruiz-Cardozo M, Vannier MW. Imaging of intranasal drug delivery to the brain. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2020; 10:1-31. [PMID: 32211216 PMCID: PMC7076302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/07/2020] [Indexed: 06/10/2023]
Abstract
Intranasal (IN) delivery is a rapidly developing area for therapies with great potential for the treatment of central nervous system (CNS) diseases. Moreover, in vivo imaging is becoming an important part of therapy assessment, both clinically in humans and translationally in animals. IN drug delivery is an alternative to systemic administration that uses the direct anatomic pathway between the olfactory/trigeminal neuroepithelium of the nasal mucosa and the brain. Several drugs have already been approved for IN application, while others are undergoing development and testing. To better understand which imaging modalities are being used to assess IN delivery of therapeutics, we performed a literature search with the key words "Intranasal delivery" and "Imaging" and summarized these findings in the current review. While this review does not attempt to be fully comprehensive, we intend for the examples provided to allow a well-rounded picture of the imaging tools available to assess IN delivery, with an emphasis on the nose-to-brain delivery route. Examples of in vivo imaging, for both humans and animals, include magnetic resonance imaging (MRI), positron emission tomography (PET), single-photon emission computed tomography (SPECT), gamma scintigraphy and computed tomography (CT). Additionally, some in vivo optical imaging modalities, including bioluminescence and fluorescence, have been used more in experimental testing in animals. In this review, we introduce each imaging modality, how it is being utilized and outline its strengths and weaknesses, specifically in the context of IN delivery of therapeutics to the brain.
Collapse
Affiliation(s)
- Michael C Veronesi
- Department of Radiology & Imaging Sciences, Indiana University School of MedicineUSA
| | - Mosa Alhamami
- Department of Radiology & Imaging Sciences, Indiana University School of MedicineUSA
| | - Shelby B Miedema
- Department of Radiology & Imaging Sciences, Indiana University School of MedicineUSA
- Department of Biomedical Engineering, Indiana University-Purdue University IndianapolisUSA
| | - Yeonhee Yun
- Department of Radiology & Imaging Sciences, Indiana University School of MedicineUSA
| | - Miguel Ruiz-Cardozo
- Clinical Research Institute, Universidad Nacional de Colombia School of MedicineUSA
| | - Michael W Vannier
- Department of Radiology, University of Chicago School of MedicineUSA
| |
Collapse
|
16
|
Hossain JA, Marchini A, Fehse B, Bjerkvig R, Miletic H. Suicide gene therapy for the treatment of high-grade glioma: past lessons, present trends, and future prospects. Neurooncol Adv 2020; 2:vdaa013. [PMID: 32642680 PMCID: PMC7212909 DOI: 10.1093/noajnl/vdaa013] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Suicide gene therapy has represented an experimental cancer treatment modality for nearly 40 years. Among the various cancers experimentally treated by suicide gene therapy, high-grade gliomas have been the most prominent both in preclinical and clinical settings. Failure of a number of promising suicide gene therapy strategies in the clinic pointed toward a bleak future of this approach for the treatment of high-grade gliomas. Nevertheless, the development of new vectors and suicide genes, better prodrugs, more efficient delivery systems, and new combinatorial strategies represent active research areas that may eventually lead to better efficacy of suicide gene therapy. These trends are evident by the current increasing focus on suicide gene therapy for high-grade glioma treatment both in the laboratory and in the clinic. In this review, we give an overview of different suicide gene therapy approaches for glioma treatment and discuss clinical trials, delivery issues, and immune responses.
Collapse
Affiliation(s)
- Jubayer A Hossain
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Haukeland University Hospital, Bergen, Norway.,Department of Oncology, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Antonio Marchini
- Department of Oncology, Luxembourg Institute of Health, Strassen, Luxembourg.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Boris Fehse
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Oncology, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
17
|
Therapeutic Efficiency of Multiple Applications of Magnetic Hyperthermia Technique in Glioblastoma Using Aminosilane Coated Iron Oxide Nanoparticles: In Vitro and In Vivo Study. Int J Mol Sci 2020; 21:ijms21030958. [PMID: 32023985 PMCID: PMC7038138 DOI: 10.3390/ijms21030958] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Magnetic hyperthermia (MHT) has been shown as a promising alternative therapy for glioblastoma (GBM) treatment. This study consists of three parts: The first part evaluates the heating potential of aminosilane-coated superparamagnetic iron oxide nanoparticles (SPIONa). The second and third parts comprise the evaluation of MHT multiple applications in GBM model, either in vitro or in vivo. The obtained heating curves of SPIONa (100 nm, +20 mV) and their specific absorption rates (SAR) stablished the best therapeutic conditions for frequencies (309 kHz and 557 kHz) and magnetic field (300 Gauss), which were stablished based on three in vitro MHT application in C6 GBM cell line. The bioluminescence (BLI) signal decayed in all applications and parameters tested and 309 kHz with 300 Gauss have shown to provide the best therapeutic effect. These parameters were also established for three MHT applications in vivo, in which the decay of BLI signal correlates with reduced tumor and also with decreased tumor glucose uptake assessed by positron emission tomography (PET) images. The behavior assessment showed a slight improvement after each MHT therapy, but after three applications the motor function displayed a relevant and progressive improvement until the latest evaluation. Thus, MHT multiple applications allowed an almost total regression of the GBM tumor in vivo. However, futher evaluations after the therapy acute phase are necessary to follow the evolution or tumor total regression. BLI, positron emission tomography (PET), and spontaneous locomotion evaluation techniques were effective in longitudinally monitoring the therapeutic effects of the MHT technique.
Collapse
|
18
|
Zhang Y, Wang WT, Gong CR, Li C, Shi M. Combination of olfactory ensheathing cells and human umbilical cord mesenchymal stem cell-derived exosomes promotes sciatic nerve regeneration. Neural Regen Res 2020; 15:1903-1911. [PMID: 32246639 PMCID: PMC7513967 DOI: 10.4103/1673-5374.280330] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Olfactory ensheathing cells (OECs) are promising seed cells for nerve regeneration. However, their application is limited by the hypoxic environment usually present at the site of injury. Exosomes derived from human umbilical cord mesenchymal stem cells have the potential to regulate the pathological processes that occur in response to hypoxia. The ability of OECs to migrate is unknown, especially in hypoxic conditions, and the effect of OECs combined with exosomes on peripheral nerve repair is not clear. Better understanding of these issues will enable the potential of OECs for the treatment of nerve injury to be addressed. In this study, OECs were acquired from the olfactory bulb of Sprague Dawley rats. Human umbilical cord mesenchymal stem cell-derived exosomes (0–400 μg/mL) were cultured with OECs for 12–48 hours. After culture with 400 μg/mL exosomes for 24 hours, the viability and proliferation of OECs were significantly increased. We observed changes to OECs subjected to hypoxia for 24 hours and treatment with exosomes. Exosomes significantly promoted the survival and migration of OECs in hypoxic conditions, and effectively increased brain-derived neurotrophic factor gene expression, protein levels and secretion. Finally, using a 12 mm left sciatic nerve defect rat model, we confirmed that OECs and exosomes can synergistically promote motor and sensory function of the injured sciatic nerve. These findings show that application of OECs and exosomes can promote nerve regeneration and functional recovery. This study was approved by the Institutional Ethical Committee of the Air Force Medical University, China (approval No. IACUC-20181004) on October 7, 2018; and collection and use of human umbilical cord specimens was approved by the Ethics Committee of the Linyi People’s Hospital, China (approval No. 30054) on May 20, 2019.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Radiation Oncology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Wen-Tao Wang
- Department of Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Chun-Rong Gong
- Rehabilitation Center, North District Hospital of the People's Hospital of Lin Yi City, Linyi, Shandong Province, China
| | - Chao Li
- Department of Orthopedics, The Eighth Medical Center of Chinese PLA general Hospital, Beijing, China
| | - Mei Shi
- Department of Radiation Oncology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
19
|
Carvalho LA, Tannous BA. Olfactory ensheathing cells travel their natural nasal pathway to deliver therapeutics to brain tumors. Oncotarget 2019; 10:4351-4353. [PMID: 31320988 PMCID: PMC6633892 DOI: 10.18632/oncotarget.27043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/10/2019] [Indexed: 11/25/2022] Open
Affiliation(s)
- Litia A Carvalho
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA, USA; Neuroscience Program, Harvard Medical School, Boston, MA, USA
| | - Bakhos A Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Neuro-Oncology Division, Massachusetts General Hospital, Boston, MA, USA; Neuroscience Program, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Lentiviral Vectors as Tools for the Study and Treatment of Glioblastoma. Cancers (Basel) 2019; 11:cancers11030417. [PMID: 30909628 PMCID: PMC6468594 DOI: 10.3390/cancers11030417] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/06/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma (GBM) has the worst prognosis among brain tumors, hence basic biology, preclinical, and clinical studies are necessary to design effective strategies to defeat this disease. Gene transfer vectors derived from the most-studied lentivirus-the Human Immunodeficiency Virus type 1-have wide application in dissecting GBM specific features to identify potential therapeutic targets. Last-generation lentiviruses (LV), highly improved in safety profile and gene transfer capacity, are also largely employed as delivery systems of therapeutic molecules to be employed in gene therapy (GT) approaches. LV were initially used in GT protocols aimed at the expression of suicide factors to induce GBM cell death. Subsequently, LV were adopted to either express small noncoding RNAs to affect different aspects of GBM biology or to overcome the resistance to both chemo- and radiotherapy that easily develop in this tumor after initial therapy. Newer frontiers include adoption of LV for engineering T cells to express chimeric antigen receptors recognizing specific GBM antigens, or for transducing specific cell types that, due to their biological properties, can function as carriers of therapeutic molecules to the cancer mass. Finally, LV allow the setting up of improved animal models crucial for the validation of GBM specific therapies.
Collapse
|