1
|
El Kheir W, Naasri S, Marcos B, Virgilio N, Paquette B, Faucheux N, Lauzon MA. CXCL12 impact on glioblastoma cells behaviors under dynamic culture conditions: Insights for developing new therapeutic approaches. PLoS One 2024; 19:e0315038. [PMID: 39715221 DOI: 10.1371/journal.pone.0315038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/19/2024] [Indexed: 12/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most prevalent malignant brain tumor, with an average survival time of 14 to 20 months. Its capacity to invade brain parenchyma leads to the failure of conventional treatments and subsequent tumor recurrence. Recent studies have explored new therapeutic strategies using a chemoattracting gradient to attract GBM cells into a soft hydrogel trap where they can be exposed to higher doses of radiation or chemotherapy. It has been demonstrated in vitro under static conditions, that nanoparticles (NPs) encapsulating the chemoattractant CXCL12 can create a gradient to attract GBM cell. However, GBM cell invasion is also largely dependent on interstitial fluid flow (IFF). In the present study, a custom-made in vitro 3D model with indirect perfusion to mimic IFF at flow rates of 0.5 μL/min and 3 μL/min was used to examine the invasive behavior of F98-rodent-derived and U87-human-derived GBM cells. This model simulated IFF and CXCL12 gradient within an alginate:matrigel-based hydrogel mimicking brain parenchyma. Findings revealed that CXCL12 (1600 ng/mL) released from NPs significantly increased the migration of F98 GBM cells after 72 hours under IFF conditions at both 0.5 and 3 μL/min. In contrast, U87 GBM cells required a higher CXCL12 concentration (2400 ng/mL) and longer incubation time for migration (120 hours). Unlike the F98 cells, U87 GBM cells showed a CXCL12 dose-dependent proliferation. Semi-quantitative qPCR showed higher CXCR4 mRNA levels in F98 cells than in U87 cells. CXCL12 significantly increased intracellular calcium levels via CXCR4 activation, with a 2.3-fold rise in F98 cells compared to U87, consistent with observed cell behavior during perfusion. This highlights the combined influence of IFF and CXCL12 on cell migration, dependent on cell line. This 3D dynamic model is a valuable tool to analyze parameters like interstitial fluid flow (IFF) and chemokine gradients, influenced by GBM tumor diversity.
Collapse
Affiliation(s)
- Wiam El Kheir
- Faculty of Engineering, Department of Chemical Engineering and Biotechnological Engineering, 3D Dynamic Cell Culture Systems Laboratory, Université de Sherbrooke, Sherbrooke, QC, Canada
- Faculty of Engineering, Department of Chemical Engineering and Biotechnological Engineering, Laboratory of Cell-Biomaterial Biohybrid Systems, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Sahar Naasri
- Faculty of Medicine and Health Sciences, Department of Medical Imaging and Radiation Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Bernard Marcos
- Faculty of Engineering, Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Nick Virgilio
- Department of Chemical Engineering, Polytechnique Montréal, Montreal, QC, Canada
| | - Benoit Paquette
- Faculty of Medicine and Health Sciences, Department of Medical Imaging and Radiation Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Clinical Research Center of the Centre Hospitalier Universitaire de l'Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Nathalie Faucheux
- Faculty of Engineering, Department of Chemical Engineering and Biotechnological Engineering, Laboratory of Cell-Biomaterial Biohybrid Systems, Université de Sherbrooke, Sherbrooke, QC, Canada
- Clinical Research Center of the Centre Hospitalier Universitaire de l'Université de Sherbrooke, Sherbrooke, QC, Canada
- The Quebec Network for Research on Protein Function, Engineering and Applications, Montreal, QC, Canada
| | - Marc-Antoine Lauzon
- Faculty of Engineering, Department of Chemical Engineering and Biotechnological Engineering, 3D Dynamic Cell Culture Systems Laboratory, Université de Sherbrooke, Sherbrooke, QC, Canada
- The Quebec Network for Research on Protein Function, Engineering and Applications, Montreal, QC, Canada
- Research Center on Aging, Sherbrooke, QC, Canada
| |
Collapse
|
2
|
Khan M, Nasim M, Feizy M, Parveen R, Gull A, Khan S, Ali J. Contemporary strategies in glioblastoma therapy: Recent developments and innovations. Neuroscience 2024; 560:211-237. [PMID: 39368608 DOI: 10.1016/j.neuroscience.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/30/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma multiforme (GBM) represents one of the most prevailing and aggressive primary brain tumors among adults. Despite advances in therapeutic approaches, the complex microenvironment of GBM poses significant challenges in its optimal therapy, which are attributed to immune evasion, tumor repopulation by stem cells, and limited drug penetration across the blood-brain barrier (BBB). Nanotechnology has emerged as a promising avenue for GBM treatment, offering biosafety, sustained drug release, enhanced solubility, and improved BBB penetrability. In this review, a comprehensive overview of recent advancements in nanocarrier-based drug delivery systems for GBM therapy is emphasized. The conventional and novel treatment modalities for GBM and the potential of nanocarriers to overcome existing limitations are comprehensively covered. Furthermore, the updates in the clinical landscape of GBM therapeutics are presented in addition to the current status of drugs and patents in the same context. Through a critical evaluation of existing literature, the therapeutic prospect and limitations of nanocarrier-based drug delivery strategies are highlighted offering insights into future research directions and clinical translation.
Collapse
Affiliation(s)
- Mariya Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Modassir Nasim
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Mohammadamin Feizy
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Rabea Parveen
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Azka Gull
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India
| | - Saba Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India.
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, Delhi, India.
| |
Collapse
|
3
|
Madeshwaran A, Vijayalakshmi P, Umapathy VR, Shanmugam R, Selvaraj C. Unlocking estrogen receptor: Structural insights into agonists and antagonists for glioblastoma therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:1-24. [PMID: 39059983 DOI: 10.1016/bs.apcsb.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Glioblastoma (GBM), a malignant brain tumor originating in glial cells, is one of the most common primary brain malignancies, affecting one in 100,000 people, typically in the frontal lobe. Estrogens, like estradiol-17 (E2), significantly influence GBM progression, metastasis, and angiogenesis. Estrogen receptors (ERs) are crucial in signal transduction and physiology, making them potential therapeutic targets. However, their roles in GBM pathogenesis remain unclear. This review explores ERs in GBM, focusing on their involvement in tumor immune evasion, modulation of the tumor microenvironment, and the mechanisms underlying GBM progression. Additionally, therapeutic opportunities targeting ERs for GBM treatment are discussed. Estrogen, synthesized primarily in ovaries and in smaller amounts by adrenal glands and fat tissues, regulates reproductive systems, bone density, skin health, and cardiovascular function. The invasive nature and heterogeneity of GBM complicate therapy development. Preclinical findings suggest that endocrine therapy with hormone receptor agonists or antagonists can extend patient survival and improve post-treatment quality of life. The ERβ pathway, in particular, shows tumor-suppressive potential, limiting glioma progression with fewer side effects. ERβ agonists could become a novel drug class for GBM treatment. Identifying biomarkers and specific therapeutic targets is crucial for early detection and improved prognosis. Estrogen and its receptors are advantageous for GBM treatment due to their regulation of numerous biological processes, ability to penetrate the blood-brain barrier, and genomic and non-genomic control of transcription, making them promising targets for GBM therapy.
Collapse
Affiliation(s)
- Asokan Madeshwaran
- Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Periyasamy Vijayalakshmi
- Department of Biotechnology and Bioinformatics, Holy Cross College (Autonomous), Tiruchirappalli, Tamil Nadu, India
| | - Vidhya Rekha Umapathy
- Department of Public Health Dentistry, Thai Moogambigai Dental College and Hospital, Chennai, Tamil Nadu, India
| | - Rajeshkumar Shanmugam
- Nano Biomedicine Lab, Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Chandrabose Selvaraj
- CsrDD LAB, Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| |
Collapse
|
4
|
Mao M, Wu Y, He Q. Recent advances in targeted drug delivery for the treatment of glioblastoma. NANOSCALE 2024; 16:8689-8707. [PMID: 38606460 DOI: 10.1039/d4nr01056f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Glioblastoma multiforme (GBM) is one of the highly malignant brain tumors characterized by significant morbidity and mortality. Despite the recent advancements in the treatment of GBM, major challenges persist in achieving controlled drug delivery to tumors. The management of GBM poses considerable difficulties primarily due to unresolved issues in the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB) and GBM microenvironment. These factors limit the uptake of anti-cancer drugs by the tumor, thus limiting the therapeutic options. Current breakthroughs in nanotechnology provide new prospects concerning unconventional drug delivery approaches for GBM treatment. Specifically, swimming nanorobots show great potential in active targeted delivery, owing to their autonomous propulsion and improved navigation capacities across biological barriers, which further facilitate the development of GBM-targeted strategies. This review presents an overview of technological progress in different drug administration methods for GBM. Additionally, the limitations in clinical translation and future research prospects in this field are also discussed. This review aims to provide a comprehensive guideline for researchers and offer perspectives on further development of new drug delivery therapies to combat GBM.
Collapse
Affiliation(s)
- Meng Mao
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Yingjie Wu
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Qiang He
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
5
|
Wei R, Li J, Lin W, Pang X, Yang H, Lai S, Wei X, Jiang X, Yuan Y, Yang R. Nanoparticle-mediated blockade of CXCL12/CXCR4 signaling enhances glioblastoma immunotherapy: Monitoring early responses with MRI radiomics. Acta Biomater 2024; 177:414-430. [PMID: 38360292 DOI: 10.1016/j.actbio.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/14/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024]
Abstract
The limited therapeutic efficacy of checkpoint blockade immunotherapy against glioblastoma is closely related to the blood-brain barrier (BBB) and tumor immunosuppressive microenvironment, where the latter is driven primarily by tumor-associated myeloid cells (TAMCs). Targeting the C-X-C motif chemokine ligand-12/C-X-C motif chemokine receptor-4 (CXCL12/CXCR4) signaling orchestrates the recruitment of TAMCs and has emerged as a promising approach for alleviating immunosuppression. Herein, we developed an iRGD ligand-modified polymeric nanoplatform for the co-delivery of CXCR4 antagonist AMD3100 and the small-molecule immune checkpoint inhibitor BMS-1. The iRGD peptide facilitated superior BBB crossing and tumor-targeting abilities both in vitro and in vivo. In mice bearing orthotopic GL261-Luc tumor, co-administration of AMD3100 and BMS-1 significantly inhibited tumor proliferation without adverse effects. A reprogramming of immunosuppression upon CXCL12/CXCR4 signaling blockade was observed, characterized by the reduction of TAMCs and regulatory T cells, and an increased proportion of CD8+T lymphocytes. The elevation of interferon-γ secreted from activated immune cells upregulated PD-L1 expression in tumor cells, highlighting the synergistic effect of BMS-1 in counteracting the PD-1/PD-L1 pathway. Finally, our research unveiled the ability of MRI radiomics to reveal early changes in the tumor immune microenvironment following immunotherapy, offering a powerful tool for monitoring treatment responses. STATEMENT OF SIGNIFICANCE: The insufficient BBB penetration and immunosuppressive tumor microenvironment greatly diminish the efficacy of immunotherapy for glioblastoma (GBM). In this study, we prepared iRGD-modified polymeric nanoparticles, loaded with a CXCR4 antagonist (AMD3100) and a small-molecule checkpoint inhibitor of PD-L1 (BMS-1) to overcome physical barriers and reprogram the immunosuppressive microenvironment in orthotopic GBM models. In this nanoplatform, AMD3100 converted the "cold" immune microenvironment into a "hot" one, while BMS-1 synergistically counteracted PD-L1 inhibition, enhancing GBM immunotherapy. Our findings underscore the potential of dual-blockade of CXCL12/CXCR4 and PD-1/PD-L1 pathways as a complementary approach to maximize therapeutic efficacy for GBM. Moreover, our study revealed that MRI radiomics provided a clinically translatable means to assess immunotherapeutic efficacy.
Collapse
Affiliation(s)
- Ruili Wei
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Department of Radiology, the Second Affiliated Hospital, South China University of Technology, Guangzhou 510180, PR China
| | - Jiamin Li
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Department of Radiology, the Second Affiliated Hospital, South China University of Technology, Guangzhou 510180, PR China
| | - Wanxian Lin
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Department of Radiology, the Second Affiliated Hospital, South China University of Technology, Guangzhou 510180, PR China
| | - Xinrui Pang
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Department of Radiology, the Second Affiliated Hospital, South China University of Technology, Guangzhou 510180, PR China
| | - Huikang Yang
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Department of Radiology, the Second Affiliated Hospital, South China University of Technology, Guangzhou 510180, PR China
| | - Shengsheng Lai
- School of Medical Equipment, Guangdong Food and Drug Vocational College, Guangzhou 510520, PR China
| | - Xinhua Wei
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Department of Radiology, the Second Affiliated Hospital, South China University of Technology, Guangzhou 510180, PR China
| | - Xinqing Jiang
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Department of Radiology, the Second Affiliated Hospital, South China University of Technology, Guangzhou 510180, PR China
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China.
| | - Ruimeng Yang
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Department of Radiology, the Second Affiliated Hospital, South China University of Technology, Guangzhou 510180, PR China.
| |
Collapse
|
6
|
Pontes B, Mendes FA. Mechanical Properties of Glioblastoma: Perspectives for YAP/TAZ Signaling Pathway and Beyond. Diseases 2023; 11:86. [PMID: 37366874 DOI: 10.3390/diseases11020086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Glioblastoma is a highly aggressive brain tumor with a poor prognosis. Recent studies have suggested that mechanobiology, the study of how physical forces influence cellular behavior, plays an important role in glioblastoma progression. Several signaling pathways, molecules, and effectors, such as focal adhesions, stretch-activated ion channels, or membrane tension variations, have been studied in this regard. Also investigated are YAP/TAZ, downstream effectors of the Hippo pathway, which is a key regulator of cell proliferation and differentiation. In glioblastoma, YAP/TAZ have been shown to promote tumor growth and invasion by regulating genes involved in cell adhesion, migration, and extracellular matrix remodeling. YAP/TAZ can be activated by mechanical cues such as cell stiffness, matrix rigidity, and cell shape changes, which are all altered in the tumor microenvironment. Furthermore, YAP/TAZ have been shown to crosstalk with other signaling pathways, such as AKT, mTOR, and WNT, which are dysregulated in glioblastoma. Thus, understanding the role of mechanobiology and YAP/TAZ in glioblastoma progression could provide new insights into the development of novel therapeutic strategies. Targeting YAP/TAZ and mechanotransduction pathways in glioblastoma may offer a promising approach to treating this deadly disease.
Collapse
Affiliation(s)
- Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Fabio A Mendes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
7
|
Tyrosine Kinase Inhibitors for Glioblastoma Multiforme: Challenges and Opportunities for Drug Delivery. Pharmaceutics 2022; 15:pharmaceutics15010059. [PMID: 36678688 PMCID: PMC9863099 DOI: 10.3390/pharmaceutics15010059] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain tumor with high mortality rates. Due to its invasiveness, heterogeneity, and incomplete resection, the treatment is very challenging. Targeted therapies such as tyrosine kinase inhibitors (TKIs) have great potential for GBM treatment, however, their efficacy is primarily limited by poor brain distribution due to the presence of the blood-brain barrier (BBB). This review focuses on the potential of TKIs in GBM therapy and provides an insight into the reasons behind unsuccessful clinical trials of TKIs in GBM despite the success in treating other cancer types. The main section is dedicated to the use of promising drug delivery strategies for targeted delivery to brain tumors. Use of brain targeted delivery strategies can help enhance the efficacy of TKIs in GBM. Among various drug delivery approaches used to bypass or cross BBB, utilizing nanocarriers is a promising strategy to augment the pharmacokinetic properties of TKIs and overcome their limitations. This is because of their advantages such as the ability to cross BBB, chemical stabilization of drug in circulation, passive or active targeting of tumor, modulation of drug release from the carrier, and the possibility to be delivered via non-invasive intranasal route.
Collapse
|
8
|
Sousa F. Brain-Targeted Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14091835. [PMID: 36145583 PMCID: PMC9500829 DOI: 10.3390/pharmaceutics14091835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 12/05/2022] Open
Affiliation(s)
- Flávia Sousa
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| |
Collapse
|