1
|
Sun S, Chen X, Ding N, Zhang M, Li X, Chen L, Sun K, Liu Y. Gamma-aminobutyric acid-mediated neuro-immune interactions in glioblastoma: Implications for prognosis and immunotherapy response. Life Sci 2024; 357:123067. [PMID: 39322177 DOI: 10.1016/j.lfs.2024.123067] [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: 03/12/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
AIMS This study aimed to investigate the role of gamma-aminobutyric acid (GABA) in the glioblastoma (GBM) tumor immune microenvironment (TIME) and its impact on prognosis and response to immunotherapy. MAIN METHODS This study employed single-cell RNA sequencing (scRNA-seq) to delineate the TIME of GBM, utilized non-negative matrix factorization (NMF) for GABA-associated cell clustering, and performed pseudotime analysis for cellular trajectories. Additionally, we integrated immunohistochemistry (IHC), immunofluorescence (IF), and protein-protein interaction (PPI) analysis to explore the regulatory mechanisms within the tumor microenvironment. KEY FINDINGS The study identified distinct GABA-associated immune cell subtypes, particularly macrophages and T-cells, with unique gene expression and developmental trajectories. The development of the GABA-associated scoring model (GABAAS), introduced novel prognostic indicators, enhancing our ability to predict patient outcomes. This study also suggests that GABA-related genes, including NDRG2 and TIMP1, play a crucial role in immune modulation, with potential implications for immunotherapy responsiveness. SIGNIFICANCE The findings underscore the potential of targeting GABA-related genes (NDRG2 and TIMP1) and M2 macrophage to reshape the glioblastoma immune landscape, offering a new frontier in personalized neuro-immunotherapy. This approach holds promise to counter individual tumor immunosuppressive mechanisms, enhancing patient outcomes.
Collapse
Affiliation(s)
- Shanyue Sun
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Xinyuan Chen
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Nannan Ding
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Miao Zhang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoru Li
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lin Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Kai Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China; College of Medical Information and Artificial Intelligence & Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Yingchao Liu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
2
|
Lu H, Wang Y, Chaudhary S, Balaga V, Ke H, Shi F, Liu J, Huo Y, Romanienko PJ, Xia B, De S, Chan CS, Shen Z. Medulloblastomas Initiated by Homologous Recombination Defects in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2007-2022. [PMID: 39168365 DOI: 10.1016/j.ajpath.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024]
Abstract
Germline mutations of homologous-recombination (HR) genes are among the top contributors to medulloblastomas. A significant portion of human medulloblastomas exhibit genomic signatures of HR defects. Whether ablation of Brca2 and Palb2, and their related Brca1 and Bccip genes, in the mouse brain can differentially initiate medulloblastomas was explored here. Conditional knockout mouse models of these HR genes and a conditional knockdown of Bccip (shBccip-KD) were established. Deletion of any of these genes led to microcephaly and neurologic defects, with Brca1- and Bccip- producing the worst defects. Trp53 co-deletion significantly rescued the microcephaly with Brca1, Palb2, and Brca2 deficiency but exhibited limited impact on Bccip- mice. For the first time, inactivation of either Brca1 or Palb2 with Trp53 was found to induce medulloblastomas. Despite shBccip-CKD being highly penetrative, Bccip/Trp53 deletions failed to induce medulloblastomas. The tumors displayed diverse immunohistochemical features and chromosome copy number variation. Although there were widespread up-regulations of cell proliferative pathways, most of the tumors expressed biomarkers of the sonic hedgehog subgroup. The medulloblastomas developed from Brca1-, Palb2-, and Brca2- mice were highly sensitive to a poly (ADP-ribose) polymerase inhibitor but not the ones from shBccip-CKD mice. These models recapitulate the spontaneous medulloblastoma development with high penetrance and a narrow time window, providing ideal platforms to test therapeutic agents with the ability to differentiate HR-defective and HR-proficient tumors.
Collapse
Affiliation(s)
- Huimei Lu
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Yuan Wang
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Shipra Chaudhary
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Varshita Balaga
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Hua Ke
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Fuqian Shi
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Jingmei Liu
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Yanying Huo
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | | | - Bing Xia
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Subhajyoti De
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Chang S Chan
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Zhiyuan Shen
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey; Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.
| |
Collapse
|
3
|
Xu J, Liu Y, Wang H, Hao J, Cao Y, Liu Z. Titanium boride nanosheets with photo-enhanced sonodynamic efficiency for glioblastoma treatment. Acta Biomater 2024; 188:344-357. [PMID: 39307260 DOI: 10.1016/j.actbio.2024.09.025] [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: 05/27/2024] [Revised: 08/20/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
Sonodynamic therapy (SDT) has garnered significant attention in cancer treatment, however, the low-yield reactive oxygen species (ROS) generation from sonosensitizers remains a major challenge. In this study, titanium boride nanosheets (TiB2 NSs) with photo-enhanced sonodynamic efficiency was fabricated for SDT of glioblastoma (GBM). Compared with commonly-used TiO2 nanoparticles, the obtained TiB2 NSs exhibited much higher ROS generation efficiency under ultrasound (US) irradiation due to their narrower band gap (2.50 eV). Importantly, TiB2 NSs displayed strong localized surface plasmon resonance (LSPR) effect in the second near-infrared (NIR II) window, which facilitated charge transfer rate and improved the separation efficiency of US-triggered electron-hole pairs, leading to photo-enhanced ROS generation efficiency. Furthermore, TiB2 NSs were encapsulated with macrophage cell membranes (CM) and then modified with RGD peptide to construct biomimetic nanoagents (TiB2@CM-RGD) for efficient blood-brain barrier (BBB) penetrating and GBM targeting. After intravenous injection into the tumor-bearing mouse, TiB2@CM-RGD can efficiently cross BBB and accumulate in the tumor sites. The tumor growth was significantly inhibited under simultaneous NIR II laser and US irradiation without causing appreciable long-term toxicity. Our work highlighted a new type of multifunctional titanium-based sonosensitizer with photo-enhanced sonodynamic efficiency for GBM treatment. STATEMENT OF SIGNIFICANCE: Titanium boride nanosheets (TiB2 NSs) with photo-enhanced sonodynamic efficiency was fabricated for SDT of glioblastoma (GBM). The obtained TiB2 NSs displayed strong localized surface plasmon resonance (LSPR) effect in the second near-infrared (NIR II) window, which facilitated charge transfer rate and improved the separation efficiency of US-triggered electron-hole pairs, leading to photo-enhanced ROS generation efficiency. Furthermore, TiB2 NSs were encapsulated with macrophage cell membranes (CM) and then modified with RGD peptide to construct biomimetic nanoagents (TiB2@CM-RGD) for efficient blood-brain barrier (BBB) penetrating and GBM targeting. After intravenous injection into the tumor-bearing mouse, TiB2@CM-RGD can efficiently cross BBB and accumulate in the tumor sites. The tumor growth was significantly inhibited under simultaneous NIR II laser and US irradiation without causing appreciable long-term toxicity.
Collapse
Affiliation(s)
- Jiaqing Xu
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China
| | - Ying Liu
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China
| | - Han Wang
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China
| | - Junxing Hao
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China
| | - Yu Cao
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China.
| | - Zhihong Liu
- College of Health Science and Engineering, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, Hubei University, Wuhan 430062, PR China.
| |
Collapse
|
4
|
Lv W, Yang F, Ge Z, Xin L, Zhang L, Zhai Y, Liu X, Guo Q, Mao X, Luo P, Zhang L, Jiang X, Zhang Y. Aberrant overexpression of myosin 1b in glioblastoma promotes angiogenesis via VEGF-myc-myosin 1b-Piezo1 axis. J Biol Chem 2024; 300:107807. [PMID: 39307302 DOI: 10.1016/j.jbc.2024.107807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive intracranial malignancy with poor prognosis. Enhanced angiogenesis is an essential hallmark of GBM, which demonstrates extensive microvascular proliferation and abnormal vasculature. Here, we uncovered the key role of myosin 1b in angiogenesis and vascular abnormality in GBM. Myosin 1b is upregulated in GBM endothelial cells (ECs) compared to the paired nonmalignant brain tissue. In our study, we found that myosin 1b promotes migration, proliferation, and angiogenesis of human/mouse brain ECs. We also found that myosin 1b expression in ECs can be regulated by vascular endothelial growth factor (VEGF) signaling through myc. Moreover, myosin 1b promotes angiogenesis via Piezo1 by enhancing Ca2+ influx, in which process VEGF can be the trigger. In conclusion, our results identified myosin 1b as a key mediator in promoting angiogenesis via mechanosensitive ion channel component 1 (Piezo1) and suggested that VEGF/myc signaling pathway could be responsible for driving the changes of myosin 1b overexpression in GBM ECs.
Collapse
Affiliation(s)
- Weifeng Lv
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fan Yang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuro-injury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Zhengmao Ge
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lele Xin
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Lingxue Zhang
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yaohong Zhai
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xian Liu
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Qingdong Guo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinggang Mao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Zhang
- China-Sweden International Joint Research Center for Brain Diseases, College of Life Sciences, Shaanxi Normal University, Xi'an, China; Jinfeng Laboratory, Chongqing, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Yanyu Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
5
|
Roshani F, Ahvar M, Ebrahimi A. Network analysis to identify driver genes and combination drugs in brain cancer. Sci Rep 2024; 14:18666. [PMID: 39134610 PMCID: PMC11319350 DOI: 10.1038/s41598-024-69705-9] [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: 04/25/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024] Open
Abstract
Brain cancer is one of the deadliest diseases, although many efforts have been made to treat it, there is no comprehensive and effective treatment approach yet. In recent years, the use of network-based analysis to identify important biological genes and pathways involved in various complex diseases, including brain cancer, has attracted the attention of researchers. The goal of this manuscript is to perform a comprehensive analysis of the various results presented related to brain cancer. For this purpose, firstly, based on the CORMINE medical database, collected all the genes related to brain cancer with a valid P-value. Then the structural and functional relationships between the above gene sets have been identified based on the STRING database. Next, in the PPI network, hub centrality analysis was performed to determine the proteins that have many connections with other proteins. After the modularization of the network, the module with the most hub vertices is considered as the most relevant module to the formation and progression of brain cancer. Since the driver vertices play an important role in biological systems, the edges of the selected module were oriented, and by analyzing the controllability of complex networks, a set of five proteins with the highest control power has been identified. Finally, based on the drug-gene interaction, a set of drugs effective on each of the driver genes has been obtained, which can potentially be used as new combination drugs. Validation of the hub and driver proteins shows that they are mainly essential proteins in the biological processes related to the various cancers and therefore the drugs that affect them can be considered as new combination therapy. The presented procedure can be used for any other complex disease.
Collapse
Affiliation(s)
| | - Mobina Ahvar
- Department of Physics, Alzahra University, Tehran, Iran
| | - Ali Ebrahimi
- Department of Physics, Alzahra University, Tehran, Iran.
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| |
Collapse
|
6
|
Charbit H, Lavon I. Investigating Expression Dynamics of miR-21 and miR-10b in Glioblastoma Cells In Vitro: Insights into Responses to Hypoxia and Secretion Mechanisms. Int J Mol Sci 2024; 25:7984. [PMID: 39063226 PMCID: PMC11277016 DOI: 10.3390/ijms25147984] [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: 06/26/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Glioblastoma poses significant challenges in oncology, with bevacizumab showing promise as an antiangiogenic treatment but with limited efficacy. microRNAs (miRNAs) 10b and 21 have emerged as potential biomarkers for bevacizumab response in glioblastoma patients. This study delves into the expression dynamics of miR-21 and miR-10b in response to hypoxia and explores their circulation mechanisms. In vitro experiments exposed glioma cells (A172, U87MG, U251) and human umbilical vein endothelial cells (HUVEC) to hypoxic conditions (1% oxygen) for 24 h, revealing heightened levels of miR-10b and miR-21 in glioblastoma cells. Manipulating miR-10b expression in U87MG, demonstrating a significant decrease in VEGF alpha (VEGFA) following miR-10b overexpression under hypoxic conditions. Size exclusion chromatography illustrated a notable shift towards miR-21 and miR-10b exosomal packaging during hypoxia. A proposed model suggests that effective bevacizumab treatment reduces VEGFA levels, heightening hypoxia and subsequently upregulating miR-21 and miR-10b expression. These miRNAs, released via exosomes, might impact various cellular processes, with miR-10b notably contributing to VEGFA level reduction. However, post-treatment increases in miR-10b and miR-21 could potentially restore cells to normoxic conditions through the downregulation of VEGF. This study highlights the intricate feedback loop involving miR-10b, miR-21, and VEGFA in glioblastoma treatment, underscoring the necessity for personalized therapeutic strategies. Further research should explore clinical implications for personalized glioma treatments.
Collapse
Affiliation(s)
| | - Iris Lavon
- Leslie and Michael Gaffin Center for Neuro-Oncology, Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah Medical Center, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| |
Collapse
|
7
|
Lee VK, Tejero R, Silvia N, Sattiraju A, Ramakrishnan A, Shen L, Wojcinski A, Kesari S, Friedel RH, Zou H, Dai G. 3D Brain Vascular Niche Model Captures Invasive Behavior and Gene Signatures of Glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.601756. [PMID: 39026692 PMCID: PMC11257506 DOI: 10.1101/2024.07.09.601756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Glioblastoma (GBM) is a lethal brain cancer with no effective treatment; understanding how GBM cells respond to tumor microenvironment remains challenging as conventional cell cultures lack proper cytoarchitecture while in vivo animal models present complexity all at once. Developing a culture system to bridge the gap is thus crucial. Here, we employed a multicellular approach using human glia and vascular cells to optimize a 3-dimensional (3D) brain vascular niche model that enabled not only long-term culture of patient derived GBM cells but also recapitulation of key features of GBM heterogeneity, in particular invasion behavior and vascular association. Comparative transcriptomics of identical patient derived GBM cells in 3D and in vivo xenotransplants models revealed that glia-vascular contact induced genes concerning neural/glia development, synaptic regulation, as well as immune suppression. This gene signature displayed region specific enrichment in the leading edge and microvascular proliferation zones in human GBM and predicted poor prognosis. Gene variance analysis also uncovered histone demethylation and xylosyltransferase activity as main themes for gene adaption of GBM cells in vivo . Furthermore, our 3D model also demonstrated the capacity to provide a quiescence and a protective niche against chemotherapy. In summary, an advanced 3D brain vascular model can bridge the gap between 2D cultures and in vivo models in capturing key features of GBM heterogeneity and unveil previously unrecognized influence of glia-vascular contact for transcriptional adaption in GBM cells featuring neural/synaptic interaction and immunosuppression.
Collapse
|
8
|
Spring BQ, Watanabe K, Ichikawa M, Mallidi S, Matsudaira T, Timerman D, Swain JWR, Mai Z, Wakimoto H, Hasan T. Red light-activated depletion of drug-refractory glioblastoma stem cells and chemosensitization of an acquired-resistant mesenchymal phenotype. Photochem Photobiol 2024. [PMID: 38922889 DOI: 10.1111/php.13985] [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: 05/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024]
Abstract
Glioblastoma stem cells (GSCs) are potent tumor initiators resistant to radiochemotherapy, and this subpopulation is hypothesized to re-populate the tumor milieu due to selection following conventional therapies. Here, we show that 5-aminolevulinic acid (ALA) treatment-a pro-fluorophore used for fluorescence-guided cancer surgery-leads to elevated levels of fluorophore conversion in patient-derived GSC cultures, and subsequent red light-activation induces apoptosis in both intrinsically temozolomide chemotherapy-sensitive and -resistant GSC phenotypes. Red light irradiation of ALA-treated cultures also exhibits the ability to target mesenchymal GSCs (Mes-GSCs) with induced temozolomide resistance. Furthermore, sub-lethal light doses restore Mes-GSC sensitivity to temozolomide, abrogating GSC-acquired chemoresistance. These results suggest that ALA is not only useful for fluorescence-guided glioblastoma tumor resection, but that it also facilitates a GSC drug-resistance agnostic, red light-activated modality to mop up the surgical margins and prime subsequent chemotherapy.
Collapse
Affiliation(s)
- Bryan Q Spring
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Physics, Northeastern University, Boston, Massachusetts, USA
| | - Kohei Watanabe
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Healthcare Optics Research Laboratory, Canon USA, Inc., Cambridge, Massachusetts, USA
| | - Megumi Ichikawa
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Tatsuyuki Matsudaira
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dmitriy Timerman
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph W R Swain
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Zhiming Mai
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hiroaki Wakimoto
- Brain Tumor Research Center and Molecular Neurosurgery Laboratory, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
9
|
Patel AS, Yanai I. A developmental constraint model of cancer cell states and tumor heterogeneity. Cell 2024; 187:2907-2918. [PMID: 38848676 PMCID: PMC11256907 DOI: 10.1016/j.cell.2024.04.032] [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: 10/02/2023] [Revised: 12/29/2023] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
Cancer is a disease that stems from a fundamental liability inherent to multicellular life forms in which an individual cell is capable of reneging on the interests of the collective organism. Although cancer is commonly described as an evolutionary process, a less appreciated aspect of tumorigenesis may be the constraints imposed by the organism's developmental programs. Recent work from single-cell transcriptomic analyses across a range of cancer types has revealed the recurrence, plasticity, and co-option of distinct cellular states among cancer cell populations. Here, we note that across diverse cancer types, the observed cell states are proximate within the developmental hierarchy of the cell of origin. We thus posit a model by which cancer cell states are directly constrained by the organism's "developmental map." According to this model, a population of cancer cells traverses the developmental map, thereby generating a heterogeneous set of states whose interactions underpin emergent tumor behavior.
Collapse
Affiliation(s)
- Ayushi S Patel
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA; Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Itai Yanai
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA; Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
10
|
Moreira R, Nóbrega C, de Almeida LP, Mendonça L. Brain-targeted drug delivery - nanovesicles directed to specific brain cells by brain-targeting ligands. J Nanobiotechnology 2024; 22:260. [PMID: 38760847 PMCID: PMC11100082 DOI: 10.1186/s12951-024-02511-7] [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: 02/15/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
Neurodegenerative diseases are characterized by extensive loss of function or death of brain cells, hampering the life quality of patients. Brain-targeted drug delivery is challenging, with a low success rate this far. Therefore, the application of targeting ligands in drug vehicles, such as lipid-based and polymeric nanoparticles, holds the promise to overcome the blood-brain barrier (BBB) and direct therapies to the brain, in addition to protect their cargo from degradation and metabolization. In this review, we discuss the barriers to brain delivery and the different types of brain-targeting ligands currently in use in brain-targeted nanoparticles, such as peptides, proteins, aptamers, small molecules, and antibodies. Moreover, we present a detailed review of the different targeting ligands used to direct nanoparticles to specific brain cells, like neurons (C4-3 aptamer, neurotensin, Tet-1, RVG, and IKRG peptides), astrocytes (Aquaporin-4, D4, and Bradykinin B2 antibodies), oligodendrocytes (NG-2 antibody and the biotinylated DNA aptamer conjugated to a streptavidin core Myaptavin-3064), microglia (CD11b antibody), neural stem cells (QTRFLLH, VPTQSSG, and NFL-TBS.40-63 peptides), and to endothelial cells of the BBB (transferrin and insulin proteins, and choline). Reports demonstrated enhanced brain-targeted delivery with improved transport to the specific cell type targeted with the conjugation of these ligands to nanoparticles. Hence, this strategy allows the implementation of high-precision medicine, with reduced side effects or unwanted therapy clearance from the body. Nevertheless, the accumulation of some of these nanoparticles in peripheral organs has been reported indicating that there are still factors to be improved to achieve higher levels of brain targeting. This review is a collection of studies exploring targeting ligands for the delivery of nanoparticles to the brain and we highlight the advantages and limitations of this type of approach in precision therapies.
Collapse
Grants
- under BrainHealth2020 projects (CENTRO-01-0145-FEDER-000008), through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalization and Portuguese national funds via FCT - Fundação para a Ciência e a Tecnologia, under projects - UIDB/04539/2020 and UIDP/04539/2020, POCI-01-0145-FEDER-030737 (NeuroStemForMJD, PTDC/BTM-ORG/30737/2017), CEECIND/04242/2017, and PhD Scholarship European Regional Development Fund (ERDF) through the Centro 2020 Regional Operational Programme
- under BrainHealth2020 projects (CENTRO-01-0145-FEDER-000008), through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalization and Portuguese national funds via FCT - Fundação para a Ciência e a Tecnologia, under projects - UIDB/04539/2020 and UIDP/04539/2020, POCI-01-0145-FEDER-030737 (NeuroStemForMJD, PTDC/BTM-ORG/30737/2017), CEECIND/04242/2017, and PhD Scholarship European Regional Development Fund (ERDF) through the Centro 2020 Regional Operational Programme
Collapse
Affiliation(s)
- Ricardo Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, polo 1, Coimbra, FMUC, 3004-504, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-504, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Clévio Nóbrega
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, 8005-139, Portugal
| | - Luís Pereira de Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, polo 1, Coimbra, FMUC, 3004-504, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-504, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal
- Institute of Interdisciplinary Research, University of Coimbra, Coimbra, 3030-789, Portugal
| | - Liliana Mendonça
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, polo 1, Coimbra, FMUC, 3004-504, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-504, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, Coimbra, 3030-789, Portugal.
| |
Collapse
|
11
|
Abousalman-Rezvani Z, Refaat A, Dehghankelishadi P, Roghani-Mamaqani H, Esser L, Voelcker NH. Insights into Targeted and Stimulus-Responsive Nanocarriers for Brain Cancer Treatment. Adv Healthc Mater 2024; 13:e2302902. [PMID: 38199238 DOI: 10.1002/adhm.202302902] [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: 08/31/2023] [Revised: 12/10/2023] [Indexed: 01/12/2024]
Abstract
Brain cancers, especially glioblastoma multiforme, are associated with poor prognosis due to the limited efficacy of current therapies. Nanomedicine has emerged as a versatile technology to treat various diseases, including cancers, and has played an indispensable role in combatting the COVID-19 pandemic as evidenced by the role that lipid nanocarrier-based vaccines have played. The tunability of nanocarrier physicochemical properties -including size, shape, surface chemistry, and drug release kinetics- has resulted in the development of a wide range of nanocarriers for brain cancer treatment. These nanocarriers can improve the pharmacokinetics of drugs, increase blood-brain barrier transfer efficiency, and specifically target brain cancer cells. These unique features would potentially allow for more efficient treatment of brain cancer with fewer side effects and better therapeutic outcomes. This review provides an overview of brain cancers, current therapeutic options, and challenges to efficient brain cancer treatment. The latest advances in nanomedicine strategies are investigated with an emphasis on targeted and stimulus-responsive nanocarriers and their potential for clinical translation.
Collapse
Affiliation(s)
- Zahra Abousalman-Rezvani
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organization, Research Way, Melbourne, VIC 3168, Australia
| | - Ahmed Refaat
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
- Pharmaceutics Department, Faculty of Pharmacy - Alexandria University, 1 El-Khartoum Square, Alexandria, 21021, Egypt
| | - Pouya Dehghankelishadi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
| | - Hossein Roghani-Mamaqani
- Faculty of Polymer Engineering, Sahand University of Technology, Tabriz, P.O. Box: 51335/1996, Iran
| | - Lars Esser
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organization, Research Way, Melbourne, VIC 3168, Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Rd, Melbourne, VIC 3168, Australia
- Department of Materials Science & Engineering, Faculty of Engineering, Monash University, 14 Alliance Ln, Melbourne, VIC 3168, Australia
| |
Collapse
|
12
|
Messina S. The RAS oncogene in brain tumors and the involvement of let-7 microRNA. Mol Biol Rep 2024; 51:531. [PMID: 38637419 PMCID: PMC11026240 DOI: 10.1007/s11033-024-09439-z] [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: 07/06/2023] [Accepted: 03/11/2024] [Indexed: 04/20/2024]
Abstract
RAS oncogenes are master regulator genes in many cancers. In general, RAS-driven cancers have an oncogenic RAS mutation that promotes disease progression (colon, lung, pancreas). In contrast, brain tumors are not necessarily RAS-driven cancers because RAS mutations are rarely observed. In particular, glioblastomas (the most lethal brain tumor) do not appear to have dominant genetic mutations that are suitable for targeted therapy. Standard treatment for most brain tumors continues to focus on maximal surgical resection, radiotherapy and chemotherapy. Yet the convergence of genomic aberrations such as EGFR, PDGFR and NF1 (some of which are clinically effective) with activation of the RAS/MAPK cascade is still considered a key point in gliomagenesis, and KRAS is undoubtedly a driving gene in gliomagenesis in mice. In cancer, microRNAs (miRNA) are small, non-coding RNAs that regulate carcinogenesis. However, the functional consequences of aberrant miRNA expression in cancer are still poorly understood. let-7 encodes an intergenic miRNA that is classified as a tumour suppressor, at least in lung cancer. Let-7 suppresses a plethora of oncogenes such as RAS, HMGA, c-Myc, cyclin-D and thus suppresses cancer development, differentiation and progression. let-7 family members are direct regulators of certain RAS family genes by binding to the sequences in their 3'untranslated region (3'UTR). let-7 miRNA is involved in the malignant behaviour in vitro-proliferation, migration and invasion-of gliomas and stem-like glioma cells as well as in vivo models of glioblastoma multiforme (GBM) via KRAS inhibition. It also increases resistance to certain chemotherapeutic agents and radiotherapy in GBM. Although let-7 therapy is not yet established, this review updates the current state of knowledge on the contribution of miRNA let-7 in interaction with KRAS to the oncogenesis of brain tumours.
Collapse
Affiliation(s)
- Samantha Messina
- Department of Science, Roma Tre University, Viale Guglielmo Marconi 446, 00146, Rome, Italy.
| |
Collapse
|
13
|
Yang F, Akhtar MN, Zhang D, El-Mayta R, Shin J, Dorsey JF, Zhang L, Xu X, Guo W, Bagley SJ, Fuchs SY, Koumenis C, Lathia JD, Mitchell MJ, Gong Y, Fan Y. An immunosuppressive vascular niche drives macrophage polarization and immunotherapy resistance in glioblastoma. SCIENCE ADVANCES 2024; 10:eadj4678. [PMID: 38416830 PMCID: PMC10901371 DOI: 10.1126/sciadv.adj4678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Cancer immunity is subjected to spatiotemporal regulation by leukocyte interaction with neoplastic and stromal cells, contributing to immune evasion and immunotherapy resistance. Here, we identify a distinct mesenchymal-like population of endothelial cells (ECs) that form an immunosuppressive vascular niche in glioblastoma (GBM). We reveal a spatially restricted, Twist1/SATB1-mediated sequential transcriptional activation mechanism, through which tumor ECs produce osteopontin to promote immunosuppressive macrophage (Mφ) phenotypes. Genetic or pharmacological ablation of Twist1 reverses Mφ-mediated immunosuppression and enhances T cell infiltration and activation, leading to reduced GBM growth and extended mouse survival, and sensitizing tumor to chimeric antigen receptor T immunotherapy. Thus, these findings uncover a spatially restricted mechanism controlling tumor immunity and suggest that targeting endothelial Twist1 may offer attractive opportunities for optimizing cancer immunotherapy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Md Naushad Akhtar
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Junyoung Shin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jay F. Dorsey
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen J. Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin D. Lathia
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Kabil MF, Badary OA, Bier F, Mousa SA, El-Sherbiny IM. A comprehensive review on lipid nanocarrier systems for cancer treatment: fabrication, future prospects and clinical trials. J Liposome Res 2024; 34:135-177. [PMID: 37144339 DOI: 10.1080/08982104.2023.2204372] [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: 05/03/2022] [Accepted: 04/02/2023] [Indexed: 05/06/2023]
Abstract
Over the last few decades, cancer has been considered a clinical challenge, being among the leading causes of mortality all over the world. Although many treatment approaches have been developed for cancer, chemotherapy is still the most utilized in the clinical setting. However, the available chemotherapeutics-based treatments have several caveats including their lack of specificity, adverse effects as well as cancer relapse and metastasis which mainly explains the low survival rate of patients. Lipid nanoparticles (LNPs) have been utilized as promising nanocarrier systems for chemotherapeutics to overcome the challenges of the currently applied therapeutic strategies for cancer treatment. Loading chemotherapeutic agent(s) into LNPs improves drug delivery at different aspects including specific targeting of tumours, and enhancing the bioavailability of drugs at the tumour site through selective release of their payload, thus reducing their undesired side effects on healthy cells. This review article delineates an overview of the clinical challenges in many cancer treatments as well as depicts the role of LNPs in achieving optimal therapeutic outcomes. Moreover, the review contains a comprehensive description of the many LNPs categories used as nanocarriers in cancer treatment to date, as well as the potential of LNPs for future applications in other areas of medicine and research.
Collapse
Affiliation(s)
- Mohamed Fawzi Kabil
- Nanomedicine Research Labs, Center for Materials Science (CMS), Zewail City of Science and Technology, Giza, Egypt
| | - Osama A Badary
- Clinical Pharmacy Department, Faculty of Pharmacy, The British University in Egypt, El-Shorouk City, Egypt
| | - Frank Bier
- AG Molekulare Bioanalytik und Bioelektronik, Institut für Biochemie und Biologie, Universität Potsdam Karl-Liebknecht-Straße 24/25, Potsdam (OT Golm), Germany
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Science (CMS), Zewail City of Science and Technology, Giza, Egypt
| |
Collapse
|
15
|
Zechel C, Loy M, Wegner C, Dahlke E, Soetje B, Baehr L, Leppert J, Ostermaier JJ, Lueg T, Nielsen J, Elßner J, Willeke V, Marzahl S, Tronnier V, Madany Mamlouk A. Molecular signature of stem-like glioma cells (SLGCs) from human glioblastoma and gliosarcoma. PLoS One 2024; 19:e0291368. [PMID: 38306361 PMCID: PMC10836714 DOI: 10.1371/journal.pone.0291368] [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: 09/20/2022] [Accepted: 08/28/2023] [Indexed: 02/04/2024] Open
Abstract
Glioblastoma multiforme (GBM) and the GBM variant gliosarcoma (GS) are among the tumors with the highest morbidity and mortality, providing only palliation. Stem-like glioma cells (SLGCs) are involved in tumor initiation, progression, therapy resistance, and relapse. The identification of general features of SLGCs could contribute to the development of more efficient therapies. Commercially available protein arrays were used to determine the cell surface signature of eight SLGC lines from GBMs, one SLGC line obtained from a xenotransplanted GBM-derived SLGC line, and three SLGC lines from GSs. By means of non-negative matrix factorization expression metaprofiles were calculated. Using the cophenetic correlation coefficient (CCC) five metaprofiles (MPs) were identified, which are characterized by specific combinations of 7-12 factors. Furthermore, the expression of several factors, that are associated with GBM prognosis, GBM subtypes, SLGC differentiation stages, or neural identity was evaluated. The investigation encompassed 24 distinct SLGC lines, four of which were derived from xenotransplanted SLGCs, and included the SLGC lines characterized by the metaprofiles. It turned out that all SLGC lines expressed the epidermal growth factor EGFR and EGFR ligands, often in the presence of additional receptor tyrosine kinases. Moreover, all SLGC lines displayed a neural signature and the IDH1 wildtype, but differed in their p53 and PTEN status. Pearson Correlation analysis identified a positive association between the pluripotency factor Sox2 and the expression of FABP7, Musashi, CD133, GFAP, but not with MGMT or Hif1α. Spherical growth, however, was positively correlated with high levels of Hif1α, CDK4, PTEN, and PDGFRβ, whereas correlations with stemness factors or MGMT (MGMT expression and promoter methylation) were low or missing. Factors highly expressed by all SLGC lines, irrespective of their degree of stemness and growth behavior, are Cathepsin-D, CD99, EMMPRIN/CD147, Intβ1, the Galectins 3 and 3b, and N-Cadherin.
Collapse
Affiliation(s)
- Christina Zechel
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
- Department of Neurosurgery, University Clinic Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Mira Loy
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Christiane Wegner
- Institute for Neuro- and Bioinformatics (INB), University Lübeck, Lübeck, Germany
| | - Eileen Dahlke
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Birga Soetje
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Laura Baehr
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Jan Leppert
- Department of Neurosurgery, University Clinic Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Johannes J. Ostermaier
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Thorben Lueg
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Jana Nielsen
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Julia Elßner
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Viktoria Willeke
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Svenja Marzahl
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Lübeck, Germany
| | - Volker Tronnier
- Department of Neurosurgery, University Clinic Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Amir Madany Mamlouk
- Institute for Neuro- and Bioinformatics (INB), University Lübeck, Lübeck, Germany
| |
Collapse
|
16
|
Rahban M, Joushi S, Bashiri H, Saso L, Sheibani V. Characterization of prevalent tyrosine kinase inhibitors and their challenges in glioblastoma treatment. Front Chem 2024; 11:1325214. [PMID: 38264122 PMCID: PMC10804459 DOI: 10.3389/fchem.2023.1325214] [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: 10/20/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive malignant primary tumor in the central nervous system. Despite extensive efforts in radiotherapy, chemotherapy, and neurosurgery, there remains an inadequate level of improvement in treatment outcomes. The development of large-scale genomic and proteomic analysis suggests that GBMs are characterized by transcriptional heterogeneity, which is responsible for therapy resistance. Hence, knowledge about the genetic and epigenetic heterogeneity of GBM is crucial for developing effective treatments for this aggressive form of brain cancer. Tyrosine kinases (TKs) can act as signal transducers, regulate important cellular processes like differentiation, proliferation, apoptosis and metabolism. Therefore, TK inhibitors (TKIs) have been developed to specifically target these kinases. TKIs are categorized into allosteric and non-allosteric inhibitors. Irreversible inhibitors form covalent bonds, which can lead to longer-lasting effects. However, this can also increase the risk of off-target effects and toxicity. The development of TKIs as therapeutics through computer-aided drug design (CADD) and bioinformatic techniques enhance the potential to improve patients' survival rates. Therefore, the continued exploration of TKIs as drug targets is expected to lead to even more effective and specific therapeutics in the future.
Collapse
Affiliation(s)
- Mahdie Rahban
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Joushi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Physiology Research Center, Institute of Neuropharmacology, Department of Physiology and Pharmacology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, Rome, Italy
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
17
|
Liu LH, Liu YF, Zhang HB, Liu XL, Zhang HW, Huang B, Lin F, Li WH. A Novel ANG-BSA/BCNU/ICG MNPs Integrated for Targeting Therapy of Glioblastoma. Technol Cancer Res Treat 2024; 23:15330338241281321. [PMID: 39444362 DOI: 10.1177/15330338241281321] [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] [Indexed: 10/25/2024] Open
Abstract
PURPOSE Develop an albumin nanoparticle-based nanoprobe for targeted glioblastoma (GBM) diagnosis and treatment, utilizing Angopep-2 for low-density lipoprotein receptor-related protein (LRP) targeting. METHODS Combined albumin-coated superparamagnetic iron oxide (SPIO), Carmustine (BCNU), and indocyanine green (ICG). Assessed morphology, size, Zeta potential, fluorescence, and drug encapsulation. Conducted in vitro fluorescence/MRI imaging and cell viability assays, and in vivo nanoprobe accumulation evaluation in brain tumors. RESULTS ANG-BSA/BCNU/ICG MNPs exhibited superior targeting and cytotoxicity against GBM cells in vitro. In vivo, enhanced brain tumor accumulation during imaging was observed. CONCLUSION This targeted imaging and drug delivery system holds promise for efficient GBM therapy and intraoperative localization, addressing Blood-brain barrier (BBB) limitations with precise drug delivery and imaging capabilities.
Collapse
Affiliation(s)
- Li-Hong Liu
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| | - Yu-Feng Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Hong-Bo Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Xiao-Lei Liu
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| | - Han-Wen Zhang
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Biao Huang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, People's Republic of China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Fan Lin
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| | - Wei-Hua Li
- Department of Radiology, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital,3002 SunGangXi Road, Shenzhen, China
| |
Collapse
|
18
|
Kumar U. Somatostatin and Somatostatin Receptors in Tumour Biology. Int J Mol Sci 2023; 25:436. [PMID: 38203605 PMCID: PMC10779198 DOI: 10.3390/ijms25010436] [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: 11/03/2023] [Revised: 12/24/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Somatostatin (SST), a growth hormone inhibitory peptide, is expressed in endocrine and non-endocrine tissues, immune cells and the central nervous system (CNS). Post-release from secretory or immune cells, the first most appreciated role that SST exhibits is the antiproliferative effect in target tissue that served as a potential therapeutic intervention in various tumours of different origins. The SST-mediated in vivo and/or in vitro antiproliferative effect in the tumour is considered direct via activation of five different somatostatin receptor subtypes (SSTR1-5), which are well expressed in most tumours and often more than one receptor in a single cell. Second, the indirect effect is associated with the regulation of growth factors. SSTR subtypes are crucial in tumour diagnosis and prognosis. In this review, with the recent development of new SST analogues and receptor-specific agonists with emerging functional consequences of signaling pathways are promising therapeutic avenues in tumours of different origins that are discussed.
Collapse
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
19
|
Levitin HM, Zhao W, Bruce JN, Canoll P, Sims PA. Consensus scHPF Identifies Cell Type-Specific Drug Responses in Glioma by Integrating Large-Scale scRNA-seq. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570193. [PMID: 38105955 PMCID: PMC10723271 DOI: 10.1101/2023.12.05.570193] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Single-cell transcriptomic analyses now frequently involve elaborate study designs including samples from multiple individuals, experimental conditions, perturbations, and batches from complex tissues. Dimensionality reduction is required to facilitate integration, interpretation, and statistical analysis. However, these datasets often include subtly different cellular subpopulations or state transitions, which are poorly described by clustering. We previously reported a Bayesian matrix factorization algorithm called single-cell hierarchical Poisson factorization (scHPF) that identifies gene co-expression patterns directly from single-cell RNA-seq (scRNA-seq) count matrices while accounting for transcript drop-out and noise. Here, we describe consensus scHPF, which analyzes scHPF models from multiple random initializations to identify the most robust gene signatures and automatically determine the number of factors for a given dataset. Consensus scHPF facilitates integration of complex datasets with highly multi-modal posterior distributions, resulting in factors that can be uniformly analyzed across individuals and conditions. To demonstrate the utility of consensus scHPF, we performed a meta-analysis of a large-scale scRNA-seq dataset from drug-treated, human glioma slice cultures generated from surgical specimens across three major cell types, 19 patients, 10 drug treatment conditions, and 52 samples. In addition to recapitulating previously reported cell type-specific drug responses from smaller studies, consensus scHPF identified disparate effects of the topoisomerase poisons etoposide and topotecan that are highly consistent with the distinct roles and expression patterns of their respective protein targets.
Collapse
Affiliation(s)
- Hanna Mendes Levitin
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Wenting Zhao
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jeffrey N Bruce
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter Canoll
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
20
|
Kumar N, Khurana B, Arora D. Nose-to-brain drug delivery for the treatment of glioblastoma multiforme: nanotechnological interventions. Pharm Dev Technol 2023; 28:1032-1047. [PMID: 37975846 DOI: 10.1080/10837450.2023.2285506] [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: 06/23/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive malignant brain tumor with a short survival rate. Extensive research is underway for the last two decades to find an effective treatment for GBM but the tortuous pathophysiology, development of chemoresistance, and presence of BBB are the major challenges, prompting scientists to look for alternative targets and delivery strategies. Therefore, the nose to brain delivery emerged as an unorthodox and non-invasive route, which delivers the drug directly to the brain via the olfactory and trigeminal pathways and also bypasses the BBB and hepatic metabolism of the drug. However, mucociliary clearance, low administration volume, and less permeability of nasal mucosa are the obstacles retrenching the brain drug concentration. Thus, nanocarrier delivery through this route may conquer these limitations because of their unique surface characteristics and smaller size. In this review, we have emphasized the advantages and limitations of nanocarrier technologies such as polymeric, lipidic, inorganic, and miscellaneous nanoparticles used for nose-to-brain drug delivery against GBM in the past 10 years. Furthermore, recent advances, patents, and clinical trials are highlighted. However, most of these studies are in the early stages, so translating their outcomes into a marketed formulation would be a milestone in the better progression and survival of glioma patients.
Collapse
Affiliation(s)
- Nitish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Bharat Khurana
- Department of Pharmaceutics, Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, India
| | - Daisy Arora
- Department of Pharmacy, Panipat Institute of Engineering and Technology, Panipat, Haryana, India
| |
Collapse
|
21
|
Kośliński P, Pluskota R, Koba M, Siedlecki Z, Śniegocki M. Comparative Analysis of Amino Acid Profiles in Patients with Glioblastoma and Meningioma Using Liquid Chromatography Electrospray Ionization Tandem Mass Spectrometry (LC-ESI-MS/MS). Molecules 2023; 28:7699. [PMID: 38067430 PMCID: PMC10707850 DOI: 10.3390/molecules28237699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Brain tumors account for 1% of all cancers diagnosed de novo. Due to the specificity of the anatomical area in which they grow, they can cause significant neurological disorders and lead to poor functional status and disability. Regardless of the results of biochemical markers of intracranial neoplasms, they are currently of no diagnostic significance. The aim of the study was to use LC-ESI-MS/MS in conjunction with multivariate statistical analyses to examine changes in amino acid metabolic profiles between patients with glioblastoma, meningioma, and a group of patients treated for osteoarthritis of the spine as a control group. Comparative analysis of amino acids between patients with glioblastoma, meningioma, and the control group allowed for the identification of statistically significant differences in the amino acid profile, including both exogenous and endogenous amino acids. The amino acids that showed statistically significant differences (lysine, histidine, α-aminoadipic acid, phenylalanine) were evaluated for diagnostic usefulness based on the ROC curve. The best results were obtained for phenylalanine. Classification trees were used to build a model allowing for the correct classification of patients into the study group (patients with glioblastoma multiforme) and the control group, in which cysteine turned out to be the most important amino acid in the decision-making algorithm. Our results indicate amino acids that may prove valuable, used alone or in combination, toward improving the diagnosis of patients with glioma and meningioma. To better assess the potential utility of these markers, their performance requires further validation in a larger cohort of samples.
Collapse
Affiliation(s)
- Piotr Kośliński
- Department of Toxicology and Bromatology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, dr. A. Jurasza 2, 85-089 Bydgoszcz, Poland; (R.P.); (M.K.)
| | - Robert Pluskota
- Department of Toxicology and Bromatology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, dr. A. Jurasza 2, 85-089 Bydgoszcz, Poland; (R.P.); (M.K.)
| | - Marcin Koba
- Department of Toxicology and Bromatology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, dr. A. Jurasza 2, 85-089 Bydgoszcz, Poland; (R.P.); (M.K.)
| | - Zygmunt Siedlecki
- Department of Neurosurgery, Neurotraumatology and Pediatric Neurosurgery, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-094 Bydgoszcz, Poland; (Z.S.); (M.Ś.)
| | - Maciej Śniegocki
- Department of Neurosurgery, Neurotraumatology and Pediatric Neurosurgery, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-094 Bydgoszcz, Poland; (Z.S.); (M.Ś.)
| |
Collapse
|
22
|
Li D, Liang Y, Yao G, Guan Z, Zhao H, Zhang N, Jiang J, Gao W. Monte Carlo-based optimization of glioma capsule design for enhanced brachytherapy. Appl Radiat Isot 2023; 201:111014. [PMID: 37688904 DOI: 10.1016/j.apradiso.2023.111014] [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: 05/11/2023] [Revised: 08/15/2023] [Accepted: 09/02/2023] [Indexed: 09/11/2023]
Abstract
The use of radiotherapy in tumor treatment has become increasingly prominent and has emerged as one of the main tools for treating malignant tumors. Current radiation therapy for glioma employs 125I seeds for brachytherapy, which cannot be combined with radiotherapy and chemotherapy. To address this limitation, this paper proposes a dual-microcavity capsule structure that integrates radiotherapy and chemotherapy. The Monte Carlo simulation method is used to simulate the structure of the dual-microcavity capsule with a 125I liquid radioactive source. Based on the simulation results, two kinds of dual-microcavity capsule structures are optimized, and the optimized dual-microcavity capsule structure is obtained. Finally, the dosimetric parameters of the two optimized dual-microcavity capsule structures are analyzed and compared with those of other 125I seeds. The optimization tests show that the improved dual-capsule dual-microcavity structure is more effective than the single-capsule dual-microcavity structure. At an activity of 5 mCi, the average absorbed dose rate is 71.2 cGy/h in the center of the optimized dual-capsule dual-microcavity structure and 45.8 cGy/h in the center of the optimized single-capsule dual-microcavity structure. Although the radial dose function and anisotropy function exhibite variations from the data of other 125I seeds, they are generally similar. The absorbed dose rate decreases exponentially with increasing distance from the center of the capsule, which can reduce the damage to the surrounding tissues and organs while increasing the dose. The capsule structure has a better irradiation effect than conventional 125I seeds and can accomplish long-term, stable, low-dose continuous irradiation to form local high-dose radiation therapy for glioma.
Collapse
Affiliation(s)
- Dongjie Li
- School of Measurement and Communication Engineering, Harbin University of Science and Technology, Harbin, China; Key Laboratory of Advanced Manufacturing and Intelligent Technology, Ministry of Education, Harbin University of Science and Technology, Harbin, China.
| | - Yu Liang
- School of Measurement and Communication Engineering, Harbin University of Science and Technology, Harbin, China; Key Laboratory of Advanced Manufacturing and Intelligent Technology, Ministry of Education, Harbin University of Science and Technology, Harbin, China
| | - Gang Yao
- Heilongjiang Institute of Atomic Energy, Harbin, China
| | - Zhongbao Guan
- Key Laboratory of Advanced Manufacturing and Intelligent Technology, Ministry of Education, Harbin University of Science and Technology, Harbin, China
| | - Hongtao Zhao
- Heilongjiang Institute of Atomic Energy, Harbin, China
| | - Nan Zhang
- Heilongjiang Institute of Atomic Energy, Harbin, China
| | - Jicheng Jiang
- Heilongjiang Institute of Atomic Energy, Harbin, China
| | - Weida Gao
- Harbin Medical University, Harbin, China
| |
Collapse
|
23
|
Phatale V, Famta P, Srinivasarao DA, Vambhurkar G, Jain N, Pandey G, Kolipaka T, Khairnar P, Shah S, Singh SB, Raghuvanshi RS, Srivastava S. Neutrophil membrane-based nanotherapeutics: Propitious paradigm shift in the management of cancer. Life Sci 2023; 331:122021. [PMID: 37582468 DOI: 10.1016/j.lfs.2023.122021] [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: 06/22/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023]
Abstract
Cancer is the leading cause of death across the globe, with 19.3 million new cancer cases and 10 million deaths in the year 2020. Conventional treatment modalities have numerous pitfalls, such as off-site cytotoxicity and poor bioavailability. Nanocarriers (NCs) have been explored to deliver various therapeutic moieties such as chemotherapeutic agents and photothermal agents, etc. However, several limitations, such as rapid clearance by the reticuloendothelial system, poor extravasation into the tumor microenvironment, and low systemic half-life are roadblocks to successful clinical translation. To circumvent the pitfalls of currently available treatment modalities, neutrophil membrane (NM)-based nanotherapeutics have emerged as a promising platform for cancer management. Their versatile features such as natural tumor tropism, tumor-specific accumulation, and prevention from rapid clearance owing to their autologous nature make them an effective anticancer NCs. In this manuscript, we have discussed various methods for isolation, coating and characterization of NM. We have discussed the role of NM-coated nanotherapeutics as neoadjuvant and adjuvant in different treatment modalities, such as chemotherapy, photothermal and photodynamic therapies with rationales behind their inclusion. Clinical hurdles faced during the bench-to-bedside translation with possible solutions have been discussed. We believe that in the upcoming years, NM-coated nanotherapeutics will open a new horizon in cancer management.
Collapse
Affiliation(s)
- Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Naitik Jain
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Tejaswini Kolipaka
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Rajeev Singh Raghuvanshi
- Central Drugs Standard Control Organization (CDSCO), Directorate General of Health Services, Ministry of Health & Family Welfare, Government of India, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
24
|
Zhang X, Shi Z, Xie Y, Wang Y, Shen C, Qi Z, Zhang L, Yang B, Yu J, Ding H. Quantitative analysis using intraoperative contrast-enhanced ultrasound in adult-type diffuse gliomas with isocitrate dehydrogenase mutations: association between hemodynamics and molecular features. Ultrasonography 2023; 42:561-571. [PMID: 37710388 PMCID: PMC10555694 DOI: 10.14366/usg.23031] [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: 02/26/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 09/16/2023] Open
Abstract
PURPOSE The relationship between contrast-enhanced ultrasound (CEUS) hemodynamics and the molecular biomarkers of adult-type diffuse gliomas, particularly isocitrate dehydrogenase (IDH), remains unclear. This study was conducted to provide a comprehensive description of the vascularization of adult-type diffuse gliomas using quantitative indicators. Additionally, it was designed to identify any variables with the potential to intraoperatively predict IDH mutation status. METHODS This prospective study enrolled patients with adult-type diffuse gliomas between November 2021 and September 2022. Intraoperative CEUS was performed, and CEUS videos were recorded for 90-second periods. Hemodynamic parameters, including the peak enhancement (PE) difference, were calculated based on the time-intensity curve of the region of interest. A differential analysis was performed on the CEUS parameters with respect to molecular biomarkers and grades. Receiver operating characteristic curves for various parameters were analyzed to evaluate the ability of those parameters to predict IDH mutation status. RESULTS Sixty patients with adult-type diffuse gliomas were evaluated. All hemodynamic parameters, apart from rising time, demonstrated significant differences between IDH-mutant and IDH-wildtype adult-type diffuse gliomas. The PE difference emerged as the optimal indicator for differentiating between IDH-wildtype and IDH-mutant gliomas, with an area under the curve of 0.958 (95% confidence interval, 0.406 to 0.785). Additionally, the hemodynamic parameters revealed significant differences across both grades and types of adult-type diffuse gliomas. CONCLUSION Hemodynamic parameters can be used intraoperatively to effectively distinguish between IDHwildtype and IDH-mutant adult-type diffuse gliomas. Additionally, quantitative CEUS equips neurosurgeons with dynamic perfusion information for various types and grades of adult-type diffuse gliomas.
Collapse
Affiliation(s)
- Xiandi Zhang
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhifeng Shi
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuanxin Xie
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Yong Wang
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Chao Shen
- Institute of Neurosurgery, Fudan University, Shanghai, China
| | - Zengxin Qi
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Liqiong Zhang
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Bojie Yang
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinhua Yu
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Hong Ding
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Borges HS, Gusmão LA, Tedesco AC. Multi-charged nanoemulsion for photodynamic treatment of glioblastoma cell line in 2D and 3D in vitro models. Photodiagnosis Photodyn Ther 2023; 43:103723. [PMID: 37487809 DOI: 10.1016/j.pdpdt.2023.103723] [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: 05/12/2023] [Revised: 07/05/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
Multi-charged nanoemulsions (NE) were designed to deliver Cannabidiol (CBD), Indocyanine green (ICG), and Protoporphyrin (PpIX) to treat glioblastoma (GBM) through Photodynamic Therapy (PDT). The phase-inversion temperature (PIT) method resulted in a highly stable NE that can be scaled easily, with a six-month shelf-life. We observed the quasi-spherical morphology of the nanoemulsions without any unencapsulated material and that 89% (± 5.5%) of the material was encapsulated. All physicochemical properties were within the expected range for a nanostructured drug delivery system, making these multi-charged nanoemulsions promising for further research and development. NE-PIC (NE-Protoporphyrin + Indocyanine + CBD) was easily internalized on GBM cells after three hours of incubation. Nanoemulsion (NE and NE-PIC) did not result in significant cytotoxicity, even for GBM or non-tumorigenic cell lines (NHF). Phototoxicity was significantly higher for the U87MG cell than the T98G cell when exposed to: visible (430 nm) and infrared (810 nm) laser light, with a difference of about 20%. From 50 mJ.cm-2, the viability of GBM cell lines decreases significantly, ranging from 65% to 85%. The NE-PIC was also effective for inhibiting cell proliferation into a 3D spheroidal GBM cell model, which is promising for mimicking the tumor cell environment. Irradiation at 810 nm was more effective in treating spheroid due to its deeper penetration in complex structures. NE-PIC has the potential as a drug delivery system for photoinactivation and photo diagnostic of GBM cell lines, taking advantage of the versatility of its active components.
Collapse
Affiliation(s)
- Hiago Salge Borges
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering ‒ Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901, Ribeirão Preto-SP, Brazil
| | - Luiza Araújo Gusmão
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering ‒ Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901, Ribeirão Preto-SP, Brazil
| | - Antonio Claudio Tedesco
- Department of Chemistry, Center of Nanotechnology and Tissue Engineering ‒ Photobiology and Photomedicine Research Group, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901, Ribeirão Preto-SP, Brazil.
| |
Collapse
|
26
|
Lu Q, Liu T, Han Z, Zhao J, Fan X, Wang H, Song J, Ye H, Sun J. Revolutionizing cancer treatment: The power of cell-based drug delivery systems. J Control Release 2023; 361:604-620. [PMID: 37579974 DOI: 10.1016/j.jconrel.2023.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/30/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Intravenous administration of drugs is a widely used cancer therapy approach. However, the efficacy of these drugs is often hindered by various biological barriers, including circulation, accumulation, and penetration, resulting in poor delivery to solid tumors. Recently, cell-based drug delivery platforms have emerged as promising solutions to overcome these limitations. These platforms offer several advantages, including prolonged circulation time, active targeting, controlled release, and excellent biocompatibility. Cell-based delivery systems encompass cell membrane coating, intracellular loading, and extracellular backpacking. These innovative platforms hold the potential to revolutionize cancer diagnosis, monitoring, and treatment, presenting a plethora of opportunities for the advancement and integration of pharmaceuticals, medicine, and materials science. Nevertheless, several technological, ethical, and financial barriers must be addressed to facilitate the translation of these platforms into clinical practice. In this review, we explore the emerging strategies to overcome these challenges, focusing specifically on the functions and advantages of cell-mediated drug delivery in cancer treatment.
Collapse
Affiliation(s)
- Qi Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Tian Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Zeyu Han
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jian Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Xiaoyuan Fan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Helin Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Jiaxuan Song
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Hao Ye
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China; Multi-Scale Robotics Lab (MSRL), Institute of Robotics & Intelligent Systems (IRIS), ETH Zurich, Zurich 8092, Switzerland.
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
27
|
Mekala JR, Adusumilli K, Chamarthy S, Angirekula HSR. Novel sights on therapeutic, prognostic, and diagnostics aspects of non-coding RNAs in glioblastoma multiforme. Metab Brain Dis 2023; 38:1801-1829. [PMID: 37249862 PMCID: PMC10227410 DOI: 10.1007/s11011-023-01234-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/09/2023] [Indexed: 05/31/2023]
Abstract
Glioblastoma Multiforme (GBM) is the primary brain tumor and accounts for 200,000 deaths each year worldwide. The standard therapy includes surgical resection followed by temozolomide (TMZ)-based chemotherapy and radiotherapy. The survival period of GBM patients is only 12-15 months. Therefore, novel treatment modalities for GBM treatment are urgently needed. Mounting evidence reveals that non-coding RNAs (ncRNAs) were involved in regulating gene expression, the pathophysiology of GBM, and enhancing therapeutic outcomes. The combinatory use of ncRNAs, chemotherapeutic drugs, and tumor suppressor gene expression induction might provide an innovative, alternative therapeutic approach for managing GBM. Studies have highlighted the role of Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) in prognosis and diagnosis. Dysregulation of ncRNAs is observed in virtually all tumor types, including GBMs. Studies have also indicated the blood-brain barrier (BBB) as a crucial factor that hinders chemotherapy. Although several nanoparticle-mediated drug deliveries were degrading effectively against GBM in vitro conditions. However, the potential to cross the BBB and optimum delivery of oligonucleotide RNA into GBM cells in the brain is currently under intense clinical trials. Despite several advances in molecular pathogenesis, GBM remains resistant to chemo and radiotherapy. Targeted therapies have less clinical benefit due to high genetic heterogeneity and activation of alternative pathways. Thus, identifying GBM-specific prognostic pathways, essential genes, and genomic aberrations provide several potential benefits as subtypes of GBM. Also, these approaches will provide insights into new strategies to overcome the heterogenous nature of GBM, which will eventually lead to successful therapeutic interventions toward precision medicine and precision oncology.
Collapse
Affiliation(s)
- Janaki Ramaiah Mekala
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram, Guntur, 522302, Andhra Pradesh, India.
| | - Kowsalya Adusumilli
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram, Guntur, 522302, Andhra Pradesh, India
| | - Sahiti Chamarthy
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram, Guntur, 522302, Andhra Pradesh, India
| | - Hari Sai Ram Angirekula
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram, Guntur, 522302, Andhra Pradesh, India
| |
Collapse
|
28
|
Shen CK, Huang BR, Charoensaensuk V, Yang LY, Tsai CF, Liu YS, Lu DY, Yeh WL, Lin C. Bradykinin B1 Receptor Affects Tumor-Associated Macrophage Activity and Glioblastoma Progression. Antioxidants (Basel) 2023; 12:1533. [PMID: 37627528 PMCID: PMC10451655 DOI: 10.3390/antiox12081533] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Bradykinin is a small active peptide and is considered an inflammatory mediator in several pathological conditions. Bradykinin exerts its effects by coupling to its receptors, including bradykinin B1 (B1R) and bradykinin B2. B1R has been implicated in the development of various cancers. Our previous study reported that B1R promoted glioblastoma (GBM) development by supporting the migration and invasion of GBM cells. However, the mechanisms underlying the effects of B1R on tumor-associated macrophages (TAMs) and GBM progression remain unknown. Accordingly, to explore the regulatory effects of B1R overexpression (OE) in GBM on tumor-associated immune cells and tumor progression, we constructed a B1R wild-type plasmid and developed a B1R OE model. The results reveal that B1R OE in GBM promoted the expression of ICAM-1 and VCAM-1-cell adhesion molecules-in GBM. Moreover, B1R OE enhanced GBM cell migration ability and monocyte attachment. B1R also regulated the production of the protumorigenic cytokines and chemokines IL-6, IL-8, CXCL11, and CCL5 in GBM, which contributed to tumor progression. We additionally noted that B1R OE in GBM increased the expression of CD68 in TAMs. Furthermore, B1R OE reduced the level of reactive oxygen species in GBM cells by upregulating heme oxygenase-1, an endogenous antioxidant protein, thereby protecting GBM cells from oxidative stress. Notably, B1R OE upregulated the expression of programmed death-ligand 1 in both GBM cells and macrophages, thus providing resistance against T-cell response. B1R OE in GBM also promoted tumor growth and reduced survival rates in an intracranial xenograft mouse model. These results indicate that B1R expression in GBM promotes TAM activity and modulates GBM progression. Therefore, B1R could be an effective target for therapeutic methods in GBM.
Collapse
Affiliation(s)
- Ching-Kai Shen
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan;
| | - Bor-Ren Huang
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427213, Taiwan
| | - Vichuda Charoensaensuk
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan (D.-Y.L.)
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Cheng-Fang Tsai
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Yu-Shu Liu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan (D.-Y.L.)
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan (D.-Y.L.)
- Department of Photonics and Communication Engineering, Asia University, Taichung 41354, Taiwan
| | - Wei-Lan Yeh
- Department of Biochemistry, School of Medicine, China Medical University, Taichung 40402, Taiwan
- Institute of New Drug Development, China Medical University, Taichung 40402, Taiwan
| | - Chingju Lin
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
29
|
Wang X, Sun Q, Wang W, Liu B, Gu Y, Chen L. Decoding key cell sub-populations and molecular alterations in glioblastoma at recurrence by single-cell analysis. Acta Neuropathol Commun 2023; 11:125. [PMID: 37525259 PMCID: PMC10391841 DOI: 10.1186/s40478-023-01613-x] [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: 04/27/2023] [Accepted: 07/01/2023] [Indexed: 08/02/2023] Open
Abstract
Glioblastoma (GBM) is the most frequent malignant brain tumor, the relapse of which is unavoidable following standard treatment. However, the effective treatment for recurrent GBM is lacking, necessitating the understanding of key mechanisms driving tumor recurrence and the identification of new targets for intervention. Here, we integrated single-cell RNA-sequencing data spanning 36 patient-matched primary and recurrent GBM (pGBM and rGBM) specimens, with 6 longitudinal GBM spatial transcriptomics to explore molecular alterations at recurrence, with each cell type characterized in parallel. Genes involved in extracellular matrix (ECM) organization are preferentially enriched in rGBM cells, and MAFK is highlighted as a potential regulator. Notably, we uncover a unique subpopulation of GBM cells that is much less detected in pGBM and highly expresses ECM and mesenchyme related genes, suggesting it may contribute to the molecular transition of rGBM. Further regulatory network analysis reveals that transcription factors, such as NFATC4 and activator protein 1 members, may function as hub regulators. All non-tumor cells alter their specific sets of genes as well and certain subgroups of myeloid cells appear to be physically associated with the mesenchyme-like GBM subpopulation. Altogether, our study provides new insights into the molecular understanding of GBM relapse and candidate targets for rGBM treatment.
Collapse
Affiliation(s)
- Xin Wang
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
- BGI Research, Hangzhou, 310030 China
| | - Qian Sun
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| | - Weiwen Wang
- China National GeneBank, BGI Research, Shenzhen, 518120 China
| | - Baohui Liu
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| | - Ying Gu
- BGI Research, Hangzhou, 310030 China
- BGI Research, Shenzhen, 518083 China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083 China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Liang Chen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
30
|
Ren AL, Wu JY, Lee SY, Lim M. Translational Models in Glioma Immunotherapy Research. Curr Oncol 2023; 30:5704-5718. [PMID: 37366911 DOI: 10.3390/curroncol30060428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/24/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023] Open
Abstract
Immunotherapy is a promising therapeutic domain for the treatment of gliomas. However, clinical trials of various immunotherapeutic modalities have not yielded significant improvements in patient survival. Preclinical models for glioma research should faithfully represent clinically observed features regarding glioma behavior, mutational load, tumor interactions with stromal cells, and immunosuppressive mechanisms. In this review, we dive into the common preclinical models used in glioma immunology, discuss their advantages and disadvantages, and highlight examples of their utilization in translational research.
Collapse
Affiliation(s)
- Alexander L Ren
- School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Janet Y Wu
- School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Si Yeon Lee
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA 94304, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA 94304, USA
| |
Collapse
|
31
|
Lab-on-a-chip systems for cancer biomarker diagnosis. J Pharm Biomed Anal 2023; 226:115266. [PMID: 36706542 DOI: 10.1016/j.jpba.2023.115266] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
Lab-on-a-chip (LOC) or micro total analysis system is one of the microfluidic technologies defined as the adaptation, miniaturization, integration, and automation of analytical laboratory procedures into a single instrument or "chip". In this article, we review developments over the past five years in the application of LOC biosensors for the detection of different types of cancer. Microfluidics encompasses chemistry and biotechnology skills and has revolutionized healthcare diagnosis. Superior to traditional cell culture or animal models, microfluidic technology has made it possible to reconstruct functional units of organs on chips to study human diseases such as cancer. LOCs have found numerous biomedical applications over the past five years, including integrated bioassays, cell analysis, metabolomics, drug discovery and delivery systems, tissue and organ physiology and disease modeling, and personalized medicine. This review provides an overview of the latest developments in microfluidic-based cancer research, with pros, cons, and prospects.
Collapse
|
32
|
Zhang D, Li AM, Hu G, Huang M, Yang F, Zhang L, Wellen KE, Xu X, Conn CS, Zou W, Kahn M, Rhoades SD, Weljie AM, Fuchs SY, Amankulor N, Yoshor D, Ye J, Koumenis C, Gong Y, Fan Y. PHGDH-mediated endothelial metabolism drives glioblastoma resistance to chimeric antigen receptor T cell immunotherapy. Cell Metab 2023; 35:517-534.e8. [PMID: 36804058 PMCID: PMC10088869 DOI: 10.1016/j.cmet.2023.01.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 10/24/2022] [Accepted: 01/26/2023] [Indexed: 02/19/2023]
Abstract
The efficacy of immunotherapy is limited by the paucity of T cells delivered and infiltrated into the tumors through aberrant tumor vasculature. Here, we report that phosphoglycerate dehydrogenase (PHGDH)-mediated endothelial cell (EC) metabolism fuels the formation of a hypoxic and immune-hostile vascular microenvironment, driving glioblastoma (GBM) resistance to chimeric antigen receptor (CAR)-T cell immunotherapy. Our metabolome and transcriptome analyses of human and mouse GBM tumors identify that PHGDH expression and serine metabolism are preferentially altered in tumor ECs. Tumor microenvironmental cues induce ATF4-mediated PHGDH expression in ECs, triggering a redox-dependent mechanism that regulates endothelial glycolysis and leads to EC overgrowth. Genetic PHGDH ablation in ECs prunes over-sprouting vasculature, abrogates intratumoral hypoxia, and improves T cell infiltration into the tumors. PHGDH inhibition activates anti-tumor T cell immunity and sensitizes GBM to CAR T therapy. Thus, reprogramming endothelial metabolism by targeting PHGDH may offer a unique opportunity to improve T cell-based immunotherapy.
Collapse
Affiliation(s)
- Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Albert M Li
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Guanghui Hu
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Menggui Huang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Department of Pathology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Crystal S Conn
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Zou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark Kahn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seth D Rhoades
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nduka Amankulor
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Yoshor
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
33
|
Khan I, Işık EB, Mahfooz S, Khan AM, Hatiboglu MA. Identification of Genetic Alterations in Rapid Progressive Glioblastoma by Use of Whole Exome Sequencing. Diagnostics (Basel) 2023; 13:diagnostics13061017. [PMID: 36980325 PMCID: PMC10047503 DOI: 10.3390/diagnostics13061017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Glioblastoma poses an inevitable threat to patients despite aggressive therapy regimes. It displays a great level of molecular heterogeneity and numerous substitutions in several genes have been documented. Next-generation sequencing techniques have identified various molecular signatures that have led to a better understanding of the molecular pathogenesis of glioblastoma. In this limited study, we sought to identify genetic variants in a small number of rare patients with aggressive glioblastoma. METHODS Five tumor tissue samples were isolated from four patients with rapidly growing glioblastoma. Genomic DNA was isolated and whole exome sequencing was used to study protein-coding regions. Generated FASTQ files were analyzed and variants were called for each sample. Variants were prioritized with different approaches and functional annotation was applied for the detrimental variants. RESULTS A total of 49,780 somatic variants were identified in the five glioblastoma samples studied, with the majority as missense substitutions. The top ten genes with the highest number of substitutions were MUC3A, MUC4, MUC6, OR4C5, PDE4DIP, AHNAK2, OR4C3, ZNF806, TTN, and RP1L1. Notably, variant prioritization after annotation indicated that the MTCH2 (Chr11: 47647265 A>G) gene sequence change was putative deleterious in all of the aggressive tumor samples. CONCLUSION The MTCH2 (Chr11: 47647265 A>G) gene substitution was identified as putative deleterious in highly aggressive glioblastomas, which merits further investigation. Moreover, a high tumor mutation burden was observed, with a signature of the highest substitutions in MUC3A, MUC4, MUC6, OR4C5, PDE4DIP, AHNAK2, OR4C3, ZNF806, TTN, and RP1L1 genes. The findings provide critical, initial data for the further rational design of genetic screening and diagnostic approaches against aggressive glioblastoma.
Collapse
Affiliation(s)
- Imran Khan
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
| | - Esra Büşra Işık
- Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
| | - Sadaf Mahfooz
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
| | - Asif M Khan
- Department of Microbiology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Damansara Heights, Kuala Lumpur 50490, Malaysia
| | - Mustafa Aziz Hatiboglu
- Department of Molecular Biology, Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakif University, Beykoz, 34820 Istanbul, Turkey
- Department of Neurosurgery, Bezmialem Vakif University Medical School, Vatan Street, Fatih, 34093 Istanbul, Turkey
| |
Collapse
|
34
|
Funakoshi Y, Sugihara Y, Uneda A, Nakashima T, Suzuki H. Recent advances in the molecular understanding of medulloblastoma. Cancer Sci 2023; 114:741-749. [PMID: 36520034 PMCID: PMC9986075 DOI: 10.1111/cas.15691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/25/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Medulloblastoma is the most common pediatric malignant brain tumor composed of four molecular subgroups. Recent intensive genomics has greatly contributed to our understanding of medulloblastoma pathogenesis. Sequencing studies identified novel mutations involved in the cyclic AMP-dependent pathway or RNA processing in the Sonic Hedgehog (SHH) subgroup, and core-binding factor subunit alpha (CBFA) complex in the group 4 subgroup. Likewise, single-cell sequencing provided detailed insights into the cell of origin associated with brain development. In this review, we will summarize recent findings by sequencing analyses for medulloblastoma.
Collapse
Affiliation(s)
- Yusuke Funakoshi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan
| | - Yuriko Sugihara
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan
| | - Atsuhito Uneda
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan
| | - Takuma Nakashima
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan
| | - Hiromichi Suzuki
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan
| |
Collapse
|
35
|
Khamis ZI, Sarker DB, Xue Y, Al-Akkary N, James VD, Zeng C, Li Y, Sang QXA. Modeling Human Brain Tumors and the Microenvironment Using Induced Pluripotent Stem Cells. Cancers (Basel) 2023; 15:cancers15041253. [PMID: 36831595 PMCID: PMC9954701 DOI: 10.3390/cancers15041253] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Brain cancer is a group of diverse and rapidly growing malignancies that originate in the central nervous system (CNS) and have a poor prognosis. The complexity of brain structure and function makes brain cancer modeling extremely difficult, limiting pathological studies and therapeutic developments. Advancements in human pluripotent stem cell technology have opened a window of opportunity for brain cancer modeling, providing a wealth of customizable methods to simulate the disease in vitro. This is achieved with the advent of genome editing and genetic engineering technologies that can simulate germline and somatic mutations found in human brain tumors. This review investigates induced pluripotent stem cell (iPSC)-based approaches to model human brain cancer. The applications of iPSCs as renewable sources of individual brain cell types, brain organoids, blood-brain barrier (BBB), and brain tumor models are discussed. The brain tumor models reviewed are glioblastoma and medulloblastoma. The iPSC-derived isogenic cells and three-dimensional (3D) brain cancer organoids combined with patient-derived xenografts will enhance future compound screening and drug development for these deadly human brain cancers.
Collapse
Affiliation(s)
- Zahraa I. Khamis
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
- High-Performance Materials Institute, Florida State University, Tallahassee, FL 32310, USA
- Laboratory of Cancer Biology and Molecular Immunology, Department of Biochemistry, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Drishty B. Sarker
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| | - Yu Xue
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| | - Nancy Al-Akkary
- Laboratory of Cancer Biology and Molecular Immunology, Department of Biochemistry, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Viviana D. James
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| | - Changchun Zeng
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
- High-Performance Materials Institute, Florida State University, Tallahassee, FL 32310, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
- Correspondence: ; Tel.: +1-850-644-8683; Fax: +1-850-644-8281
| |
Collapse
|
36
|
Yang F, Zhang D, Jiang H, Ye J, Zhang L, Bagley SJ, Winkler J, Gong Y, Fan Y. Small-molecule toosendanin reverses macrophage-mediated immunosuppression to overcome glioblastoma resistance to immunotherapy. Sci Transl Med 2023; 15:eabq3558. [PMID: 36791206 PMCID: PMC10394757 DOI: 10.1126/scitranslmed.abq3558] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 01/17/2023] [Indexed: 02/17/2023]
Abstract
T cell-based immunotherapy holds promise for treating solid tumors, but its therapeutic efficacy is limited by intratumoral immune suppression. This immune suppressive tumor microenvironment is largely driven by tumor-associated myeloid cells, including macrophages. Here, we report that toosendanin (TSN), a small-molecule compound, reprograms macrophages to enforce antitumor immunity in glioblastoma (GBM) in mouse models. Our functional screen of genetically probed macrophages with a chemical library identifies that TSN reverses macrophage-mediated tumor immunosuppression, leading to enhanced T cell infiltration, activation, and reduced exhaustion. Chemoproteomic and structural analyses revealed that TSN interacts with Hck and Lyn to abrogate suppressive macrophage immunity. In addition, a combination of immune checkpoint blockade and TSN therapy induced regression of syngeneic GBM tumors in mice. Furthermore, TSN treatment sensitized GBM to Egfrviii chimeric antigen receptor (CAR) T cell therapy. These findings suggest that TSN may serve as a therapeutic compound that blocks tumor immunosuppression and circumvents tumor resistance to T cell-based immunotherapy in GBM and other solid tumors that warrants further investigation.
Collapse
Affiliation(s)
- Fan Yang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Haowen Jiang
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen J. Bagley
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffery Winkler
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
37
|
Meco D, Attinà G, Mastrangelo S, Navarra P, Ruggiero A. Emerging Perspectives on the Antiparasitic Mebendazole as a Repurposed Drug for the Treatment of Brain Cancers. Int J Mol Sci 2023; 24:ijms24021334. [PMID: 36674870 PMCID: PMC9862092 DOI: 10.3390/ijms24021334] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/02/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Repurposing approved non-antitumor drugs is a promising and affordable strategy in drug discovery to identify new therapeutic uses different from the original medical indication that may help increase the number of possible, effective anticancer drugs. The use of drugs in ways other than their original FDA-approved indications could offer novel avenues such as bypassing the chemoresistance and recurrence seen with conventional therapy and treatment; moreover, it can offer a safe and economic strategy for combination therapy. Recent works have demonstrated the anticancer properties of the FDA-approved drug Mebendazole. This synthetic benzimidazole proved effective against a broad spectrum of intestinal Helminthiasis. Mebendazole can penetrate the blood-brain barrier and has been shown to inhibit the malignant progression of glioma by targeting signaling pathways related to cell proliferation, apoptosis, or invasion/migration, or by increasing the sensitivity of glioma cells to conventional chemotherapy or radiotherapy. Moreover, several preclinical models and ongoing clinical trials explore the efficacy of Mebendazole in multiple cancers, including acute myeloid leukemia, brain cancer, oropharyngeal squamous cell carcinoma, breast cancer, gastrointestinal cancer, lung carcinoma, adrenocortical carcinoma, prostate cancer, and head and neck cancer. The present review summarizes central literature regarding the anticancer effects of MBZ in cancer cell lines, animal tumor models, and clinical trials to suggest possible strategies for safe and economical combinations of anticancer therapies in brain cancer. Mebendazole might be an excellent candidate for the treatment of brain tumors because of its efficacy both when used as monotherapy and in combination as an enhancement to standard chemotherapeutics and radiotherapy, due to its effectiveness on tumor angiogenesis inhibition, cell cycle arrest, apoptosis induction, and targeting of critical pathways involved in cancer such as Hedgehog signaling. Therefore, attention to MBZ repurposing has recently increased because of its potential therapeutic versatility and significant clinical implications, such as reducing medical care costs and optimizing existing therapies. Using new treatments is essential, particularly when current therapeutics for patients with brain cancer fail.
Collapse
Affiliation(s)
- Daniela Meco
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giorgio Attinà
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Pierluigi Navarra
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-06-3058203; Fax: +39-06-3052751
| |
Collapse
|
38
|
Katsushima K, Joshi K, Perera RJ. Diagnostic and therapeutic potential of circular RNA in brain tumors. Neurooncol Adv 2023; 5:vdad063. [PMID: 37334165 PMCID: PMC10276536 DOI: 10.1093/noajnl/vdad063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023] Open
Abstract
Circular RNAs (circRNAs) are a class of RNA with a stable cyclic structure. They are expressed in various tissues and cells with conserved, specific characteristics. CircRNAs have been found to play critical roles in a wide range of cellular processes by regulating gene expression at the epigenetic, transcriptional, and posttranscriptional levels. There is an accumulation of evidence on newly discovered circRNAs, their molecular interactions, and their roles in the development and progression of human brain tumors, including cell proliferation, cell apoptosis, invasion, and chemoresistance. Here we summarize the current state of knowledge of the circRNAs that have been implicated in brain tumor pathogenesis, particularly in gliomas and medulloblastomas. In providing a comprehensive overview of circRNA studies, we highlight how different circRNAs have oncogenic or tumor-suppressive roles in brain tumors, making them attractive therapeutic targets and biomarkers for personalized therapy and precision diagnostics. This review article discusses circRNAs' functional roles and the prospect of using them as diagnostic biomarkers and therapeutic targets in patients with brain tumors.
Collapse
Affiliation(s)
- Keisuke Katsushima
- Department of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Cancer and Blood Disorders Institute, Johns Hopkins All Children’s Hospital, Florida, USA
| | - Kandarp Joshi
- Department of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Cancer and Blood Disorders Institute, Johns Hopkins All Children’s Hospital, Florida, USA
| | - Ranjan J Perera
- Corresponding Author: Ranjan J. Perera, PhD, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, 1650 Orleans St., Baltimore, MD 21231, USA ()
| |
Collapse
|
39
|
Barzegar-Fallah A, Gandhi K, Rizwan SB, Slatter TL, Reynolds JNJ. Harnessing Ultrasound for Targeting Drug Delivery to the Brain and Breaching the Blood–Brain Tumour Barrier. Pharmaceutics 2022; 14:pharmaceutics14102231. [PMID: 36297666 PMCID: PMC9607160 DOI: 10.3390/pharmaceutics14102231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Despite significant advances in developing drugs to treat brain tumours, achieving therapeutic concentrations of the drug at the tumour site remains a major challenge due to the presence of the blood–brain barrier (BBB). Several strategies have evolved to enhance brain delivery of chemotherapeutic agents to treat tumours; however, most approaches have several limitations which hinder their clinical utility. Promising studies indicate that ultrasound can penetrate the skull to target specific brain regions and transiently open the BBB, safely and reversibly, with a high degree of spatial and temporal specificity. In this review, we initially describe the basics of therapeutic ultrasound, then detail ultrasound-based drug delivery strategies to the brain and the mechanisms by which ultrasound can improve brain tumour therapy. We review pre-clinical and clinical findings from ultrasound-mediated BBB opening and drug delivery studies and outline current therapeutic ultrasound devices and technologies designed for this purpose.
Collapse
Affiliation(s)
- Anita Barzegar-Fallah
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Kushan Gandhi
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Shakila B. Rizwan
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
- School of Pharmacy, University of Otago, Dunedin 9016, New Zealand
| | - Tania L. Slatter
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - John N. J. Reynolds
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
- Correspondence: ; Tel.: +64-3-479-5781; Fax: +64-3-479-7254
| |
Collapse
|
40
|
Hesse F, Wright AJ, Somai V, Bulat F, Kreis F, Brindle KM. Imaging Glioblastoma Response to Radiotherapy Using 2H Magnetic Resonance Spectroscopy Measurements of Fumarate Metabolism. Cancer Res 2022; 82:3622-3633. [PMID: 35972377 PMCID: PMC9530651 DOI: 10.1158/0008-5472.can-22-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/25/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022]
Abstract
Early detection of tumor cell death in glioblastoma following treatment with chemoradiation has the potential to distinguish between true disease progression and pseudoprogression. Tumor cell death can be detected noninvasively in vivo by imaging the production of [2,3-2H2]malate from [2,3-2H2]fumarate using 2H magnetic resonance (MR) spectroscopic imaging. We show here that 2H MR spectroscopy and spectroscopic imaging measurements of [2,3-2H2]fumarate metabolism can detect tumor cell death in orthotopically implanted glioblastoma models within 48 hours following the completion of chemoradiation. Following the injection of [2,3-2H2]fumarate into tumor-bearing mice, production of [2,3-2H2]malate was measured in a human cell line-derived model and in radiosensitive and radioresistant patient-derived models of glioblastoma that were treated with temozolomide followed by targeted fractionated irradiation. The increase in the [2,3-2H2]malate/[2,3-2H2]fumarate signal ratio posttreatment, which correlated with histologic assessment of cell death, was a more sensitive indicator of treatment response than diffusion-weighted and contrast agent-enhanced 1H MRI measurements, which have been used clinically to detect responses of glioblastoma to chemoradiation. Overall, early detection of glioblastoma cell death using 2H MRI of malate production from fumarate could help improve the clinical evaluation of response to chemoradiation. SIGNIFICANCE 2H magnetic resonance imaging of labeled fumarate metabolism can detect early evidence of tumor cell death following chemoradiation, meeting a clinical need to reliably detect treatment response in glioblastoma.
Collapse
Affiliation(s)
- Friederike Hesse
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Alan J. Wright
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Vencel Somai
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Radiology, School of Clinical Medicine, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Flaviu Bulat
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Felix Kreis
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kevin M. Brindle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
41
|
Peng B, Hao S, Tong Z, Bai H, Pan S, Lim KL, Li L, Voelcker NH, Huang W. Blood-brain barrier (BBB)-on-a-chip: a promising breakthrough in brain disease research. LAB ON A CHIP 2022; 22:3579-3602. [PMID: 36004771 DOI: 10.1039/d2lc00305h] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The blood-brain barrier (BBB) represents a key challenge in developing brain-penetrating therapeutic molecules. BBB dysfunction is also associated with the onset and progression of various brain diseases. The BBB-on-a-chip (μBBB), an organ-on-chip technology, has emerged as a powerful in vitro platform that closely mimics the human BBB microenvironments. While the μBBB technology has seen wide application in the study of brain cancer, its utility in other brain disease models ("μBBB+") is less appreciated. Based on the advances of the μBBB technology and the evolution of in vitro models for brain diseases over the last decade, we propose the concept of a "μBBB+" system and summarize its major promising applications in pathological studies, personalized medical research, drug development, and multi-organ-on-chip approaches. We believe that such a sophisticated "μBBB+" system is a highly tunable and promising in vitro platform for further advancement of the understanding of brain diseases.
Collapse
Affiliation(s)
- Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China.
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Shiping Hao
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Ziqiu Tong
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Sijun Pan
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, Fujian, China
| | - Kah-Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, 308232, Singapore
| | - Lin Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China.
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211800, China
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, Fujian, China
| | - Nicolas H Voelcker
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China.
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China.
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211800, China
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, Fujian, China
| |
Collapse
|
42
|
Genomic and Epigenomic Features of Glioblastoma Multiforme and its Biomarkers. JOURNAL OF ONCOLOGY 2022; 2022:4022960. [PMID: 36185622 PMCID: PMC9519330 DOI: 10.1155/2022/4022960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/14/2022] [Accepted: 09/10/2022] [Indexed: 11/22/2022]
Abstract
Glioblastoma multiforme is a serious and life-threatening tumor of central nervous system, characterized by aggressive behavior, poor prognosis, and low survival rate. Despite of the availability of aggressive antitumor therapeutic regimen for glioblastoma (radiotherapy followed by chemotherapeutic dose), recovery rate, and patients' survival ratio is attributed to the lack of selectivity of therapeutic drugs and less advancement in cancer therapeutics over last decade. Moreover, tools employed in conventional diagnosis of glioblastoma are more invasive and painful, making the process excruciating for the patients. These challenges urge for the need of novel biomarkers for diagnosis, prognosis, and prediction purpose with less invasiveness and more patient compliance. This article will explore the genetic biomarkers isocitrate dehydrogenase mutation, MGMT mutations, and EGFR that can be deployed as an analytical tool in diagnosis of disease and prognosis of a therapeutic course. The review also highlights the importance of employing novel microRNAs as prognostic biomarkers. Recent clinical advancements to treat GBM and to prevent relapse of the disease are also discussed in this article in the hope of finding a robust and effective method to treat GBM.
Collapse
|
43
|
Xiong A, Zhang J, Chen Y, Zhang Y, Yang F. Integrated single-cell transcriptomic analyses reveal that GPNMB-high macrophages promote PN-MES transition and impede T cell activation in GBM. EBioMedicine 2022; 83:104239. [PMID: 36054938 PMCID: PMC9437813 DOI: 10.1016/j.ebiom.2022.104239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 10/27/2022] Open
|
44
|
Wang J, Le W, Yan T, Jiang J, Chen B. Usage of Nanoparticles to Alter Neutrophils' Function for Therapy. ACS Biomater Sci Eng 2022; 8:3676-3689. [PMID: 36018296 DOI: 10.1021/acsbiomaterials.2c00711] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Neutrophils, the most abundant white blood cells in the human body, are important immune cells responsible for the innate immune response. Neutrophils can migrate to inflammatory areas, such as tumor sites and infection sites, because of chemotaxis. Neutrophil-based nanomaterials, such as neutrophil-nanomaterial composites and neutrophil membrane-based nanomaterials, can help the drug or imaging agent gather in the inflammatory area with the help of chemotaxis. In addition, some nanomaterials can interfere with the function of neutrophils to treat tissue damage caused by excessive local accumulation of neutrophils. This review focuses on the interaction between nanomaterials and neutrophils as well as the applications of neutrophil-based nanomaterials and neutrophil-interfering nanomaterials.
Collapse
Affiliation(s)
- Jing Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai 200092, China
| | - Wenjun Le
- Shanghai East Hospital Ji'an Hospital, 80 Ji'an South Road, Ji'an City 343000, Jiangxi Province, China
| | - Tinghua Yan
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinhua Jiang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bingdi Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
45
|
A Novel Thrombosis-Related Signature for Predicting Survival and Drug Compounds in Glioblastoma. JOURNAL OF ONCOLOGY 2022; 2022:6792850. [PMID: 35874629 PMCID: PMC9300384 DOI: 10.1155/2022/6792850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/12/2022] [Indexed: 12/02/2022]
Abstract
Glioblastoma is the most common primary tumor in the central nervous system, and thrombosis-associated genes are related to its occurrence and progression. Univariate Cox and LASSO regression analysis were utilized to develop a new prognostic signature based on thrombosis-associated genes. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and HALLMARK were used for functional annotation of risk signature. ESTIMATE, MCP-counter, xCell, and TIMER algorithms were used to quantify immune infiltration in the tumor microenvironment. Genomics of Drug Sensitivity in Cancer (GDSC) was used for selecting potential drug compounds. Risk signature based on thrombosis-associated genes shows moderate performance in prognosis prediction. The functional annotation of the risk signature indicates that the signaling pathways related to the cell cycle, apoptosis, tumorigenesis, and immune suppression are rich in the high-risk group. Somatic mutation analysis shows that tumor-suppressive gene TP53 and oncogene PTEN have higher expression in low-risk and high-risk groups, respectively. Potential drug compounds are explored in risk score groups and show higher AUC values in the low-risk score group. A nomogram with valuable prognostic factors exhibits high sensitivity in predicting the survival outcome of GBM patients. Our research screens out multiple thromboses-associated genes with remarkable clinical significance in GBM and further develops a meaningful prognostic risk signature predicting drug sensitivity and survival outcome.
Collapse
|
46
|
Role of Circular RNA in Brain Tumor Development. Cells 2022; 11:cells11142130. [PMID: 35883576 PMCID: PMC9315629 DOI: 10.3390/cells11142130] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022] Open
Abstract
Central nervous system tumors are a leading cause of cancer-related death in children and adults, with medulloblastoma (MB) and glioblastoma (GBM) being the most prevalent malignant brain tumors, respectively. Despite tremendous breakthroughs in neurosurgery, radiation, and chemotherapeutic techniques, cell heterogeneity and various genetic mutations impacting cell cycle control, cell proliferation, apoptosis, and cell invasion result in unwanted resistance to treatment approaches, with a 5-year survival rate of 70–80% for medulloblastoma, and the median survival time for patients with glioblastoma is only 15 months. Developing new medicines and utilizing combination medications may be viewed as excellent techniques for battling MB and GBM. Circular RNAs (circRNAs) can affect cancer-developing processes such as cell proliferation, cell apoptosis, invasion, and chemoresistance in this regard. As a result, several compounds have been introduced as prospective therapeutic targets in the fight against MB and GBM. The current study aims to elucidate the fundamental molecular and cellular mechanisms underlying the pathogenesis of GBM in conjunction with circRNAs. Several mechanisms were examined in detail, including PI3K/Akt/mTOR signaling, Wnt/-catenin signaling, angiogenic processes, and metastatic pathways, in order to provide a comprehensive knowledge of the involvement of circRNAs in the pathophysiology of MB and GBM.
Collapse
|
47
|
Perspective on the Use of DNA Repair Inhibitors as a Tool for Imaging and Radionuclide Therapy of Glioblastoma. Cancers (Basel) 2022; 14:cancers14071821. [PMID: 35406593 PMCID: PMC8997380 DOI: 10.3390/cancers14071821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 01/03/2023] Open
Abstract
Simple Summary The current routine treatment for glioblastoma (GB), the most lethal high-grade brain tumor in adults, aims to induce DNA damage in the tumor. However, the tumor cells might be able to repair that damage, which leads to therapy resistance. Fortunately, DNA repair defects are common in GB cells, and their survival is often based on a sole backup repair pathway. Hence, targeted drugs inhibiting essential proteins of the DNA damage response have gained momentum and are being introduced in the clinic. This review gives a perspective on the use of radiopharmaceuticals targeting DDR kinases for imaging in order to determine the DNA repair phenotype of GB, as well as for effective radionuclide therapy. Finally, four new promising radiopharmaceuticals are suggested with the potential to lead to a more personalized GB therapy. Abstract Despite numerous innovative treatment strategies, the treatment of glioblastoma (GB) remains challenging. With the current state-of-the-art therapy, most GB patients succumb after about a year. In the evolution of personalized medicine, targeted radionuclide therapy (TRT) is gaining momentum, for example, to stratify patients based on specific biomarkers. One of these biomarkers is deficiencies in DNA damage repair (DDR), which give rise to genomic instability and cancer initiation. However, these deficiencies also provide targets to specifically kill cancer cells following the synthetic lethality principle. This led to the increased interest in targeted drugs that inhibit essential DDR kinases (DDRi), of which multiple are undergoing clinical validation. In this review, the current status of DDRi for the treatment of GB is given for selected targets: ATM/ATR, CHK1/2, DNA-PK, and PARP. Furthermore, this review provides a perspective on the use of radiopharmaceuticals targeting these DDR kinases to (1) evaluate the DNA repair phenotype of GB before treatment decisions are made and (2) induce DNA damage via TRT. Finally, by applying in-house selection criteria and analyzing the structural characteristics of the DDRi, four drugs with the potential to become new therapeutic GB radiopharmaceuticals are suggested.
Collapse
|
48
|
Zhang J, Guan M, Zhou X, Berry K, He X, Lu QR. Long Noncoding RNAs in CNS Myelination and Disease. Neuroscientist 2022; 29:287-301. [PMID: 35373640 DOI: 10.1177/10738584221083919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Myelination by oligodendrocytes is crucial for neuronal survival and function, and defects in myelination or failure in myelin repair can lead to axonal degeneration and various neurological diseases. At present, the factors that promote myelination and overcome the remyelination block in demyelinating diseases are poorly defined. Although the roles of protein-coding genes in oligodendrocyte differentiation have been extensively studied, the majority of the mammalian genome is transcribed into noncoding RNAs, and the functions of these molecules in myelination are poorly characterized. Long noncoding RNAs (lncRNAs) regulate transcription at multiple levels, providing spatiotemporal control and robustness for cell type-specific gene expression and physiological functions. lncRNAs have been shown to regulate neural cell-type specification, differentiation, and maintenance of cell identity, and dysregulation of lncRNA function has been shown to contribute to neurological diseases. In this review, we discuss recent advances in our understanding of the functions of lncRNAs in oligodendrocyte development and myelination as well their roles in neurological diseases and brain tumorigenesis. A more systematic characterization of lncRNA functional networks will be instrumental for a better understanding of CNS myelination, myelin disorders, and myelin repair.
Collapse
Affiliation(s)
- Jing Zhang
- Laboratory of Nervous System Injuries and Diseases, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children at Sichuan University, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Menglong Guan
- Laboratory of Nervous System Injuries and Diseases, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children at Sichuan University, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xianyao Zhou
- Laboratory of Nervous System Injuries and Diseases, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children at Sichuan University, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Kalen Berry
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xuelian He
- Laboratory of Nervous System Injuries and Diseases, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children at Sichuan University, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
49
|
Liu SH, Wang QZ, Liu T, Bai R, Ma MM, Liu QL, Zhou HG, Liu J, Wang M. Enhanced Glioblastoma Selectivity of Harmine via the Albumin Carrier. J Biomed Nanotechnol 2022; 18:1052-1063. [PMID: 35854453 DOI: 10.1166/jbn.2022.3321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Glioblastoma, the most common tumor in the brain, has witnessed very little clinical progress over the last decades. Exploring and discovering new therapeutic strategies for glioblastoma has become a critical problem. Harmine (HM), belonging to the beta-carboline alkaloid, is a natural product and isolated from the seeds of Peganum harmala L., which own notable antitumor activity in vitro. However, the poor water solubility and less selectivity of HM severely limit its clinical use. For enhancing its selective ability to tumor cells, we fabricated a kind of protein nanoparticles (BSA-HM NPs), composed of the modified bovine serum albumin (BSA) and HM. It was substantiated through in vitro and in vivo experiment that BSA-HM NPs could predominantly accumulate in tumor tissues and exhibited remarkably enhanced antitumor efficacy. This study provides a promising strategy to improve the bioavailability and avoid side effects of HM as antitumor agents by choosing BSA as carriers.
Collapse
Affiliation(s)
- Shi-Hui Liu
- Department of Pharmaceutics, College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, P. R. China
| | - Qing-Zhen Wang
- Department of Pharmaceutics, College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, P. R. China
| | - Tao Liu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Ru Bai
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Man-Man Ma
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Qiao-Lin Liu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Hui-Ge Zhou
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Jing Liu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and Center for Excellence in Nanoscience, Chinese Academy of Sciences, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Mei Wang
- Department of Pharmaceutics, College of Pharmacy, Xinjiang Medical University, Urumqi, 830054, P. R. China
| |
Collapse
|
50
|
Wu T, Liu Y, Cao Y, Liu Z. Engineering Macrophage Exosome Disguised Biodegradable Nanoplatform for Enhanced Sonodynamic Therapy of Glioblastoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110364. [PMID: 35133042 DOI: 10.1002/adma.202110364] [Citation(s) in RCA: 126] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/26/2022] [Indexed: 06/14/2023]
Abstract
Sonodynamic therapy (SDT) exhibits high tissue penetration and negligible radiation damage to normal tissues, and thus emerges as a promising cancer therapeutic modality for glioblastoma (GBM). However, the blood-brain barrier (BBB) and hypoxic microenvironment greatly limit the SDT efficiency. In this work, a biodegradable nanoplatform (termed as CSI) is fabricated by encapsulating catalase (CAT) into silica nanoparticles (CAT@SiO2 ) for tumor hypoxia relief, and then loaded with the sonosensitizer indocyanine green (ICG). Inspired by the ability of macrophages to cross the BBB, CSI is further coated with AS1411 aptamer-modified macrophage exosomes to form CSI@Ex-A, which possesses efficient BBB penetration and good cancer-cell-targeting capability. After tumor cell endocytosis, highly expressed glutathione (GSH) triggeres biodegradation of the nanoplatform and the released CAT catalyzes hydrogen peroxide (H2 O2 ) to produce O2 to relieve tumor hypoxia. The GSH depletion and O2 self-supplying effectively enhances the SDT efficiency both in vitro and in vivo. In addition, the resulting CSI@Ex-A exhibits good biocompatibility and long circulation time. These findings demonstrate that CSI@Ex-A may serve as a competent nanoplatform for GBM therapy, with potential for clinical translation.
Collapse
Affiliation(s)
- Tingting Wu
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, P. R. China
| | - Ying Liu
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, P. R. China
| | - Yu Cao
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, P. R. China
| | - Zhihong Liu
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, P. R. China
| |
Collapse
|