1
|
Janowicz PW, Boele T, Maschmeyer RT, Gholami YH, Kempe EG, Stringer BW, Stoner SP, Zhang M, du Toit-Thompson T, Williams F, Touffu A, Munoz L, Kuncic Z, Brighi C, Waddington DEJ. Enhanced detection of glioblastoma vasculature with superparamagnetic iron oxide nanoparticles and MRI. Sci Rep 2025; 15:14283. [PMID: 40274951 DOI: 10.1038/s41598-025-97943-y] [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: 01/22/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Detecting glioblastoma infiltration in the brain is challenging due to limited MRI contrast beyond the enhancing tumour core. This study aims to investigate the potential of superparamagnetic iron oxide nanoparticles (SPIONs) as contrast agents for improved detection of diffuse brain cancer. We examine the distribution and pharmacokinetics of SPIONs in glioblastoma models with intact and disrupted blood-brain barriers. Using MRI, we imaged RN1-luc and U87MG mice injected with Gadovist and SPIONs, observing differences in blood-brain barrier permeability. Peripheral imaging showed strong uptake of nanoparticles in the liver and spleen, while vascular and renal signals were transient. Susceptibility gradient mapping enabled positive nanoparticle contrast within tumours and provided additional information on tumour angiogenesis. This approach offers a novel method for detecting diffuse brain cancer. Our findings demonstrate that SPIONs enhance glioblastoma detection beyond conventional MRI, providing insights into tumour angiogenesis and opening new avenues for early diagnosis and targeted treatment strategies.
Collapse
Affiliation(s)
- Phillip W Janowicz
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| | - Thomas Boele
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Richard T Maschmeyer
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
| | - Yaser H Gholami
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, Australia
| | - Emma G Kempe
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Brett W Stringer
- Institute for Biomedicine and Glycomics, Griffith University, Brisbane, Australia
| | - Shihani P Stoner
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Marie Zhang
- Imagion Biosystems Ltd, Melbourne, Australia
| | - Taymin du Toit-Thompson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Fern Williams
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Aude Touffu
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Lenka Munoz
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Zdenka Kuncic
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
- The Sydney Nano Institute, The University of Sydney, Sydney, Australia
| | - Caterina Brighi
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - David E J Waddington
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
2
|
Wu Y, Hao C, Gao C, Hageman M, Lee S, Kirkland TA, Gray NS, Su Y, Lin MZ. Pharmacodynamics of Akt drugs revealed by a kinase-modulated bioluminescent indicator. Nat Chem Biol 2025:10.1038/s41589-025-01846-y. [PMID: 39934397 DOI: 10.1038/s41589-025-01846-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
Measuring pharmacodynamics (PD)-the biochemical effects of drug dosing-and correlating them with therapeutic efficacy in animal models is crucial for the development of effective drugs but traditional PD studies are labor and resource intensive. Here we developed a kinase-modulated bioluminescent indicator (KiMBI) for rapid, noninvasive PD assessment of Akt-targeted drugs, minimizing drug and animal use. Using KiMBI, we performed a structure-PD relationship analysis on the brain-active Akt inhibitor ipatasertib by generating and characterizing two novel analogs. One analog, ML-B01, successfully inhibited Akt in both the brain and the body. Interestingly, capivasertib, ipatasertib and ML-B01 all exhibited PD durations beyond their pharmacokinetic profiles. Furthermore, KiMBI revealed that the PD effects of an Akt-targeted proteolysis-targeting chimera degrader endured for over 3 days. Thus, bioluminescence imaging with Akt KiMBI provides a noninvasive and efficient method for in vivo visualization of the PD of Akt inhibitors and degraders.
Collapse
Affiliation(s)
- Yan Wu
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Chenzhou Hao
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | - Chao Gao
- Promega Corporation, San Luis Obispo, CA, USA
| | | | - Sungmoo Lee
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | | | - Nathanael S Gray
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Yichi Su
- Department of Nuclear Medicine, Zhongshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Michael Z Lin
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Department of Neurobiology, Stanford University, Stanford, CA, USA.
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA.
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
3
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
4
|
Jin T, Li B, Li L, Qi W, Xi L. High spatiotemporal mapping of cortical blood flow velocity with an enhanced accuracy. BIOMEDICAL OPTICS EXPRESS 2024; 15:2419-2432. [PMID: 38633086 PMCID: PMC11019678 DOI: 10.1364/boe.520886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 04/19/2024]
Abstract
Cerebral blood flow velocity is one of the most essential parameters related to brain functions and diseases. However, most existing mapping methods suffer from either inaccuracy or lengthy sampling time. In this study, we propose a particle-size-related calibration method to improve the measurement accuracy and a random-access strategy to suppress the sampling time. Based on the proposed methods, we study the long-term progress of cortical vasculopathy and abnormal blood flow caused by glioma, short-term variations of blood flow velocity under different anesthetic depths, and cortex-wide connectivity of the rapid fluctuation of blood flow velocities during seizure onset. The experimental results demonstrate that the proposed calibration method and the random-access strategy can improve both the qualitative and quantitative performance of velocimetry techniques and are also beneficial for understanding brain functions and diseases from the perspective of cerebral blood flow.
Collapse
Affiliation(s)
- Tian Jin
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Baochen Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Linyang Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Weizhi Qi
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Lei Xi
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
5
|
Zhao F, Wang X, Zhu W, Zhao D, Ye C, Guo Y, Dou Y. Low-dose pleiotropic radiosensitive nanoformulations for three-pronged radiochemotherapy of hypoxic brain glioblastoma under BOLD/DWI monitoring. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00159-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
Background
Hypoxia-mediated radioresistance is the main obstacle to the successful treatment of glioblastoma (GBM). Enhancing hypoxic radiosensitivity and alleviating tumor hypoxia are both effective means to improve therapeutic efficacy, and the combination of the two is highly desirable and meaningful.
Results
Herein, we construct a low-dose pleiotropic radiosensitive nanoformulation consisting of a high-Z atomic nanocrystal core and mesoporous silica shell, surface-modified with angiopep-2 (ANG) peptide and loaded with nitric oxide (NO) donor and hypoxia-activated prodrug (AQ4N). Benefiting from ANG-mediated transcytosis, this nanoformulation can efficiently cross the BBB and accumulate preferentially in the brain. Low-dose radiation triggers this nanoformulation to exert a three-pronged synergistic therapeutic effect through high-Z-atom-dependent dose deposition enhancement, NO-mediated hypoxia relief, and AQ4N-induced hypoxia-selective killing, thereby significantly inhibiting GBM in situ growth while prolonging survival and maintaining stable body weight in the glioma-bearing mice. Meanwhile, the proposed in vivo 9.4 T BOLD/DWI can realize real-time dynamic assessment of local oxygen supply and radiosensitivity to monitor the therapeutic response of GBM.
Conclusions
This work provides a promising alternative for hypoxia-specific GBM-targeted comprehensive therapy, noninvasive monitoring, and precise prognosis.
Graphical Abstract
Collapse
|
6
|
Padmakumar S, Amiji MM. Long-Acting Therapeutic Delivery Systems for the Treatment of Gliomas. Adv Drug Deliv Rev 2023; 197:114853. [PMID: 37149040 DOI: 10.1016/j.addr.2023.114853] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
Despite the emergence of cutting-edge therapeutic strategies and tremendous progress in research, a complete cure of glioma remains elusive. The heterogenous nature of tumor, immunosuppressive state and presence of blood brain barrier are few of the major obstacles in this regard. Long-acting depot formulations such as injectables and implantables are gaining attention for drug delivery to brain owing to their ease in administration and ability to elute drug locally for extended durations in a controlled manner with minimal toxicity. Hybrid matrices fabricated by incorporating nanoparticulates within such systems help to enhance pharmaceutical advantages. Utilization of long-acting depots as monotherapy or in conjunction with existing strategies rendered significant survival benefits in many preclinical studies and some clinical trials. The discovery of novel targets, immunotherapeutic strategies and alternative drug administration routes are now coupled with several long-acting systems with an ultimate aim to enhance patient survival and prevent glioma recurrences.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, 02115; Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, 02115.
| |
Collapse
|
7
|
Wu Y, Walker JR, Westberg M, Ning L, Monje M, Kirkland TA, Lin MZ, Su Y. Kinase-Modulated Bioluminescent Indicators Enable Noninvasive Imaging of Drug Activity in the Brain. ACS CENTRAL SCIENCE 2023; 9:719-732. [PMID: 37122464 PMCID: PMC10141594 DOI: 10.1021/acscentsci.3c00074] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Indexed: 05/03/2023]
Abstract
Aberrant kinase activity contributes to the pathogenesis of brain cancers, neurodegeneration, and neuropsychiatric diseases, but identifying kinase inhibitors that function in the brain is challenging. Drug levels in blood do not predict efficacy in the brain because the blood-brain barrier prevents entry of most compounds. Rather, assessing kinase inhibition in the brain requires tissue dissection and biochemical analysis, a time-consuming and resource-intensive process. Here, we report kinase-modulated bioluminescent indicators (KiMBIs) for noninvasive longitudinal imaging of drug activity in the brain based on a recently optimized luciferase-luciferin system. We develop an ERK KiMBI to report inhibitors of the Ras-Raf-MEK-ERK pathway, for which no bioluminescent indicators previously existed. ERK KiMBI discriminates between brain-penetrant and nonpenetrant MEK inhibitors, reveals blood-tumor barrier leakiness in xenograft models, and reports MEK inhibitor pharmacodynamics in native brain tissues and intracranial xenografts. Finally, we use ERK KiMBI to screen ERK inhibitors for brain efficacy, identifying temuterkib as a promising brain-active ERK inhibitor, a result not predicted from chemical characteristics alone. Thus, KiMBIs enable the rapid identification and pharmacodynamic characterization of kinase inhibitors suitable for treating brain diseases.
Collapse
Affiliation(s)
- Yan Wu
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
| | - Joel R. Walker
- Promega
Biosciences LLC, San Luis Obispo, California 93401, United States
| | - Michael Westberg
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
- Department
of Chemistry, Aarhus University, Aarhus 8000, Denmark
| | - Lin Ning
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
| | - Michelle Monje
- Department
of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, United States
- Howard Hughes
Medical Institute, Stanford University, Stanford, California 94305, United States
| | - Thomas A. Kirkland
- Promega
Biosciences LLC, San Luis Obispo, California 93401, United States
| | - Michael Z. Lin
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
- Department
of Pediatrics, Stanford University, Stanford, California 94305, United States
- Department
of Chemical and Systems Biology, Stanford
University, Stanford, California 94305, United States
| | - Yichi Su
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
8
|
Qiao R, Fu C, Forgham H, Javed I, Huang X, Zhu J, Whittaker AK, Davis TP. Magnetic Iron Oxide Nanoparticles for Brain Imaging and Drug Delivery. Adv Drug Deliv Rev 2023; 197:114822. [PMID: 37086918 DOI: 10.1016/j.addr.2023.114822] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/14/2023] [Accepted: 04/09/2023] [Indexed: 04/24/2023]
Abstract
Central nervous system (CNS) disorders affect as many as 1.5 billion people globally. The limited delivery of most imaging and therapeutic agents into the brain is a major challenge for treatment of CNS disorders. With the advent of nanotechnologies, controlled delivery of drugs with nanoparticles holds great promise in CNS disorders for overcoming the blood-brain barrier (BBB) and improving delivery efficacy. In recent years, magnetic iron oxide nanoparticles (MIONPs) have stood out as a promising theranostic nanoplatform for brain imaging and drug delivery as they possess unique physical properties and biodegradable characteristics. In this review, we summarize the recent advances in MIONP-based platforms as imaging and drug delivery agents for brain diseases. We firstly introduce the methods of synthesis and surface functionalization of MIONPs with emphasis on the inclusion of biocompatible polymers that allow for the addition of tailored physicochemical properties. We then discuss the recent advances in in vivo imaging and drug delivery applications using MIONPs. Finally, we present a perspective on the remaining challenges and possible future directions for MIONP-based brain delivery systems.
Collapse
Affiliation(s)
- Ruirui Qiao
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Changkui Fu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Helen Forgham
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ibrahim Javed
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Xumin Huang
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jiayuan Zhu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andrew K Whittaker
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Thomas P Davis
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
9
|
Nicola JP, LaRocca CJ. Sodium iodide symporter-targeted gene therapy in glioblastoma. Mol Ther Oncolytics 2023; 28:44-45. [PMID: 36654785 PMCID: PMC9827349 DOI: 10.1016/j.omto.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Juan Pablo Nicola
- Department of Clinical Biochemistry, Faculty of Chemical Sciences, National University of Córdoba, Córdoba, Argentina,Clinical Biochemistry and Immunology Research Center - National Scientific and Technical Research Council (CIBICI-CONICET), Córdoba, Argentina,Corresponding author: Juan Pablo Nicola, Department of Clinical Biochemistry, Faculty of Chemical Sciences, National University of Córdoba, Córdoba, Argentina.
| | - Christopher J. LaRocca
- Department of Surgery, Division of Surgical Oncology, University of Minnesota, Minneapolis, MN, USA,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA,Corresponding author: Christopher J. LaRocca, Department of Surgery, Division of Surgical Oncology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
10
|
Rani V, Prabhu A. In vitro blood brain barrier models: Molecular aspects and therapeutic strategies in glioma management. Curr Res Transl Med 2023; 71:103376. [PMID: 36580825 DOI: 10.1016/j.retram.2022.103376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Glioma management is the most challenging task in clinical oncology due to numerous reasons. One of the major hurdles in glioma therapy is the presence of blood brain barrier which resists the entry of most of the drugs into the brain. However, in case of tumors, blood brain barrier integrity is compromised, which in turn can be advantageous in delivering the drugs, if the therapeutic module is strategically modified. For such improvised therapeutic strategy, it is necessary to understand the molecular composition and profiling of blood brain barrier and blood brain tumor barrier. This review mainly focuses on the composition, markers expressed on the blood brain barrier which will help the readers to understand its basic environment. It also gives a detailed account of the various in vitro models that are used to study the nature of the blood brain barrier and describes various strategies in improvising the drug delivery in glioma management.
Collapse
Affiliation(s)
- Vinitha Rani
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, 575018 Karnataka, India
| | - Ashwini Prabhu
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, 575018 Karnataka, India.
| |
Collapse
|
11
|
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: 16] [Impact Index Per Article: 5.3] [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
|
12
|
Yin L, Yao Z, Wang Y, Mazuranic M. Investigational cyclin-dependent kinase 4/6 inhibitor GLR2007 demonstrates activity against isocitrate dehydrogenase wild-type glioblastoma and other solid tumors in mice xenograft models. Front Oncol 2022; 12:915862. [PMID: 36033522 PMCID: PMC9403987 DOI: 10.3389/fonc.2022.915862] [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: 04/08/2022] [Accepted: 07/21/2022] [Indexed: 11/18/2022] Open
Abstract
Cyclin-dependent kinases, CDK4 and CDK6, are essential in regulating the cell cycle, which is disrupted in cancers like isocitrate dehydrogenase wild-type glioblastoma (GBM). Currently marketed CDK4/6 inhibitors, including abemaciclib, have shown preclinical efficacy in solid tumors, but factors such as poor blood–brain barrier (BBB) penetration limit their efficacy in GBM. GLR2007 is an investigational CDK4/6 inhibitor with the potential for improved BBB penetration. In vitro assays were used to assess the potency and inhibition of CDK4/6 enzymatic activity of GLR2007. Using in vivo assays, the distribution of radiolabeled GLR2007 in rats was determined through quantitative whole-body autoradiography. The antitumor efficacy of GLR2007 was evaluated in human GBM and breast cancer orthotopic mice xenograft models, and human lung, colorectal, and liver cancer in a subcutaneous xenograft model. In tumor cell line proliferation assays, GLR2007 inhibited proliferation at lower concentration values than abemaciclib in 19 of 20 GBM, five of seven breast, 20 of 21 lung, and 24 of 24 liver cancer cell lines. Total levels of radiolabeled GLR2007 in the brains of rats exceeded those in plasma by 2.3–4.5-fold from 2–6 hours after dosing. A xenograft model showed that, compared with vehicle control, 50 mg/kg GLR2007 induced 95.9% tumor growth inhibition (TGI) (P<0.001) in GBM orthotopic xenografts, 81.4% TGI (P=0.037) in breast cancer orthotopic xenografts, and 91.5% TGI (P<0.001) in colorectal cancer subcutaneous xenografts. These studies show possible BBB penetration of GLR2007 and demonstrate its potential as a CDK4/6 inhibitor for the treatment of solid tumors, including GBM.
Collapse
Affiliation(s)
- Lei Yin
- Gan & Lee Pharmaceuticals, Beijing, China
- *Correspondence: Lei Yin,
| | | | - Yue Wang
- Gan & Lee Pharmaceuticals USA Corp., Bridgewater, NJ, United States
| | | |
Collapse
|
13
|
Ma K, Chen X, Zhao X, Chen S, Yang J. PLVAP is associated with glioma-associated malignant processes and immunosuppressive cell infiltration as a promising marker for prognosis. Heliyon 2022; 8:e10298. [PMID: 36033326 PMCID: PMC9404362 DOI: 10.1016/j.heliyon.2022.e10298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 04/25/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Previous reports have confirmed the significance of plasmalemma vesicle-associated protein (PLVAP) in the progression of multiple tumors; however, there are few studies examining its immune properties in the context of gliomas. Here, we methodically investigated the pathophysiological characteristics and clinical manifestations of gliomas. A total of 699 patients diagnosed with gliomas in the cancer genome atlas along with 325 glioma patients in the Chinese glioma genome atlas were collected for the training and validation sets. We analyzed and visualized the total statistics using RStudio. PLVAP was markedly upregulated among high grade gliomas, O6-methylguanine-DNA methyltransferase promoter unmethylated subforms, isocitrate dehydrogenase wild forms, 1p19q non-codeletion subforms, and mesenchyme type gliomas. The receiver operating characteristics analysis illustrated the favorable applicability of PLVAP in regard to estimating mesenchyme subform gliomas. Subsequent Kaplan–Meier curves together with multivariable Cox analyses upon survival identified high-expression PLVAP as a distinct prognostic variable for patients with gliomas. Gene ontology analysis of PLVAP among gliomas has documented the predominant role of this protein in glioma-associated immunobiological processes and also in inflammatory responses. We consequently examined the associations of PLVAP with immune-related meta-genes, and PLVAP was positively correlated with hematopoietic cell kinase, lymphocyte-specific protein tyrosine kinase, major histocompatibility complex (MHC) I, MHC II, signal transducer and activator of transcription 1, and interferon and was negatively correlated with immunoglobulin G. Moreover, association analyses between PLVAP and glioma-infiltrating immunocytes indicated that the infiltrating degrees of most immune cells exhibited positive correlations with PLVAP expression, particularly immunosuppressive subsets such as tumor-related macrophages, myeloid-derived suppressor cells, and regulatory T lymphocytes. In summary, we originally demonstrated that PLVAP is markedly associated with immunosuppressive immune cell infiltration degrees, unfavorable survival, and adverse pathology types among gliomas, thus identifying PLVAP as a practicable marker and a promising target for glioma-based precise diagnosis and therapeutic strategies.
Collapse
Affiliation(s)
- Kaiming Ma
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Xin Chen
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China.,Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| | - Xiaofang Zhao
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Suhua Chen
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Jun Yang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China.,Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing, China
| |
Collapse
|
14
|
Wang S, Chen L, Feng Y, Yin T, Yu J, De Keyzer F, Peeters R, Van Ongeval C, Bormans G, Swinnen J, Soete J, Wevers M, Li Y, Ni Y. Development and characterization of a rat brain metastatic tumor model by multiparametric magnetic resonance imaging and histomorphology. Clin Exp Metastasis 2022; 39:479-493. [PMID: 35218457 DOI: 10.1007/s10585-022-10155-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/07/2022] [Indexed: 02/06/2023]
Abstract
To facilitate the development of new brain metastasis (BM) treatment, an easy-to-use and clinically relevant animal model with imaging platform is needed. Rhabdomyosarcoma BM was induced in WAG/Rij rats. Post-implantation surveillance and characterizations were systematically performed with multiparametric MRI including 3D T1 and T2 weighted imaging, diffusion-weighted imaging (DWI), T1 and T2 mapping, and perfusion-weighted imaging (PWI), which were validated by postmortem digital radiography (DR), µCT angiography and histopathology. The translational potential was exemplified by the application of a vascular disrupting agent (VDA). BM was successfully induced in most rats of both genders (18/20). Multiparametric MRI revealed significantly higher T2 value, pre-contrast-enhanced (preCE) T1 value, DWI-derived apparent diffusion coefficient (ADC) and CE ratio, but a lower post-contrast-enhanced (postCE) T1 value in BM lesions than in adjacent brain (p < 0.01). PWI showed the dynamic and higher contrast agent uptake in the BM compared with the adjacent brain. DR, µCT and histopathology characterized the BM as hypervascular tumors. After VDA treatment, the BM showed drug-related perfusion changes and partial necrosis as evidenced by anatomical, functional MRI parameters and postmortem findings. The present BM model and imaging modalities represent a feasible and translational platform for developing BM-targeting therapeutics.
Collapse
Affiliation(s)
- Shuncong Wang
- KU Leuven, Biomedical Group, Campus Gasthuisberg, 3000, Leuven, Belgium
| | - Lei Chen
- KU Leuven, Biomedical Group, Campus Gasthuisberg, 3000, Leuven, Belgium
| | - Yuanbo Feng
- KU Leuven, Biomedical Group, Campus Gasthuisberg, 3000, Leuven, Belgium
| | - Ting Yin
- KU Leuven, Biomedical Group, Campus Gasthuisberg, 3000, Leuven, Belgium.,MR Collaborations, Siemens Healthineers Ltd, Shanghai, China
| | - Jie Yu
- KU Leuven, Biomedical Group, Campus Gasthuisberg, 3000, Leuven, Belgium
| | - Frederik De Keyzer
- Department of Radiology, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Ronald Peeters
- Department of Radiology, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Chantal Van Ongeval
- Department of Radiology, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Guy Bormans
- KU Leuven, Biomedical Group, Campus Gasthuisberg, 3000, Leuven, Belgium
| | - Johan Swinnen
- KU Leuven, Biomedical Group, Campus Gasthuisberg, 3000, Leuven, Belgium
| | - Jeroen Soete
- KU Leuven, Department of Materials Engineering, Kasteelpark Arenberg 44, 3001, Leuven, Belgium
| | - Martine Wevers
- KU Leuven, Department of Materials Engineering, Kasteelpark Arenberg 44, 3001, Leuven, Belgium
| | - Yue Li
- KU Leuven, Biomedical Group, Campus Gasthuisberg, 3000, Leuven, Belgium. .,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| | - Yicheng Ni
- KU Leuven, Biomedical Group, Campus Gasthuisberg, 3000, Leuven, Belgium. .,Department of Radiology, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
15
|
Gao X, Xu J, Yao T, Liu X, Zhang H, Zhan C. Peptide-decorated nanocarriers penetrating the blood-brain barrier for imaging and therapy of brain diseases. Adv Drug Deliv Rev 2022; 187:114362. [PMID: 35654215 DOI: 10.1016/j.addr.2022.114362] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
Abstract
Blood-Brain Barrier (BBB) is one of the most important physiological barriers strictly restricting the substance exchange between blood and brain tissues. While the BBB protects the brain from infections and toxins and maintains brain homeostasis, it is also recognized as the main obstacle to the penetration of therapeutics and imaging agents into the brain. Due to high specificity and affinity, peptides are frequently exploited to decorate nanocarriers across the BBB for diagnosis and/or therapy purposes. However, there are still some challenges that restrict their clinical application, such as stability, safety and immunocompatibility. In this review, we summarize the biological and pathophysiological characteristics of the BBB, strategies across the BBB, and recent progress on peptide decorated nanocarriers for brain diseases diagnosis and therapy. The challenges and opportunities for their translation are also discussed.
Collapse
|
16
|
Translation of focused ultrasound for blood-brain barrier opening in glioma. J Control Release 2022; 345:443-463. [PMID: 35337938 DOI: 10.1016/j.jconrel.2022.03.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022]
Abstract
Survival outcomes for patients with glioblastoma multiforme (GBM) have remained poor for the past 15 years, reflecting a clear challenge in the development of more effective treatment strategies. The efficacy of systemic therapies for GBM is greatly limited by the presence of the blood-brain barrier (BBB), which prevents drug penetration and accumulation in regions of infiltrative tumour, as represented in a consistent portion of GBM lesions. Focused ultrasound (FUS) - a technique that uses low-frequency ultrasound waves to induce targeted temporary disruption of the BBB - promises to improve survival outcomes by enhancing drug delivery and accumulation to infiltrating tumour regions. In this review we discuss the current state of preclinical investigations using FUS to enhance delivery of systemic therapies to intracranial neoplasms. We highlight critical methodological inconsistencies that are hampering clinical translation of FUS and we provide guiding principles for future preclinical studies. Particularly, we focus our attention on the importance of the selection of clinically relevant animal models and to the standardization of methods for FUS delivery, which will be paramount to the successful clinical translation of this promising technology for treatment in GBM patients. We also discuss how preclinical FUS research can benefit the development of GBM immunotherapies.
Collapse
|
17
|
Quader S, Kataoka K, Cabral H. Nanomedicine for brain cancer. Adv Drug Deliv Rev 2022; 182:114115. [PMID: 35077821 DOI: 10.1016/j.addr.2022.114115] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 12/18/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023]
Abstract
CNS tumors remain among the deadliest forms of cancer, resisting conventional and new treatment approaches, with mortality rates staying practically unchanged over the past 30 years. One of the primary hurdles for treating these cancers is delivering drugs to the brain tumor site in therapeutic concentration, evading the blood-brain (tumor) barrier (BBB/BBTB). Supramolecular nanomedicines (NMs) are increasingly demonstrating noteworthy prospects for addressing these challenges utilizing their unique characteristics, such as improving the bioavailability of the payloadsviacontrolled pharmacokinetics and pharmacodynamics, BBB/BBTB crossing functions, superior distribution in the brain tumor site, and tumor-specific drug activation profiles. Here, we review NM-based brain tumor targeting approaches to demonstrate their applicability and translation potential from different perspectives. To this end, we provide a general overview of brain tumor and their treatments, the incidence of the BBB and BBTB, and their role on NM targeting, as well as the potential of NMs for promoting superior therapeutic effects. Additionally, we discuss critical issues of NMs and their clinical trials, aiming to bolster the potential clinical applications of NMs in treating these life-threatening diseases.
Collapse
Affiliation(s)
- Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 212-0821, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| |
Collapse
|
18
|
Abstract
In 2001, the concept of the neurovascular unit was introduced at the Stroke Progress Review Group meeting. The neurovascular unit is an important element of the health and disease status of blood vessels and nerves in the central nervous system. Since then, the neurovascular unit has attracted increasing interest from research teams, who have contributed greatly to the prevention, treatment, and prognosis of stroke and neurodegenerative diseases. However, additional research is needed to establish an efficient, low-cost, and low-energy in vitro model of the neurovascular unit, as well as enable noninvasive observation of neurovascular units in vivo and in vitro. In this review, we first summarize the composition of neurovascular units, then investigate the efficacy of different types of stem cells and cell culture methods in the construction of neurovascular unit models, and finally assess the progress of imaging methods used to observe neurovascular units in recent years and their positive role in the monitoring and investigation of the mechanisms of a variety of central nervous system diseases.
Collapse
Affiliation(s)
- Taiwei Dong
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Min Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Feng Gao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Peifeng Wei
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Jian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Provinve, China
| |
Collapse
|
19
|
Zhao P, Anami Y, Gao P, Fan X, Li L, Tsuchikama K, Zhang N, An Z. Enhanced anti-angiogenetic effect of transferrin receptor-mediated delivery of VEGF-trap in a glioblastoma mouse model. MAbs 2022; 14:2057269. [PMID: 35388745 PMCID: PMC8993059 DOI: 10.1080/19420862.2022.2057269] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is a common and aggressive brain cancer that accounts for 60% of adult brain tumors. Anti-angiogenesis therapy is an attractive option due to the high vasculature density of GBM. However, the best-known anti-angiogenic therapeutics, bevacizumab, and aflibercept, have failed to show significant benefits in GBM patients. One of the reasons is the limited brain penetration of antibody-based therapies due to existence of the blood-brain barrier (BBB), which is further strengthened by the blood vessel normalization effects induced by anti-angiogenic therapies. To investigate if increased drug concentration in the brain by transferrin receptor (TfR)-mediated delivery across the BBB can enhance efficacy of anti-angiogenic antibody therapies, we first identified an antibody that binds to the apical domain of the mouse TfR and does not compete with the natural ligand transferrin (Tf) binding to TfR. Then, we engineered two bispecific antibodies fusing a vascular endothelial growth factor (VEGF)-Trap with the TfR-targeting antibody. Characterization of the two bispecific formats using multiple in vitro assays, which include endocytosis, cell surface and whole-cell TfR levels, human umbilical vein endothelial cell growth inhibition, and binding affinity, demonstrated that the VEGF-Trap fused with a monovalent αTfR (VEGF-Trap/moAb4) has desirable endocytosis without the induction of TfR degradation. Peripherally administered VEGF-Trap/moAb4 improved the brain concentration of VEGF-Trap by more than 10-fold in mice. The distribution of VEGF-Trap/moAb4 was validated to be in the brain parenchyma, indicating the molecule was not trapped inside the vasculature. Moreover, improved VEGF-Trap brain distribution significantly inhibited the angiogenesis of U-87 MG GBM tumors in a mouse model.
Collapse
Affiliation(s)
- Peng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yasuaki Anami
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Peng Gao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xuejun Fan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Leike Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Kyoji Tsuchikama
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, the University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
20
|
Adaptation of laser interstitial thermal therapy for tumor ablation under MRI monitoring in a rat orthotopic model of glioblastoma. Acta Neurochir (Wien) 2021; 163:3455-3463. [PMID: 34554269 DOI: 10.1007/s00701-021-05002-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/01/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Laser interstitial thermal therapy (LITT) under magnetic resonance imaging (MRI) monitoring is being increasingly used in cytoreductive surgery of recurrent brain tumors and tumors located in eloquent brain areas. The objective of this study was to adapt this technique to an animal glioma model. METHODS A rat model of U251 glioblastoma (GBM) was employed. Tumor location and extent were determined by MRI and dynamic contrast-enhanced (DCE) MRI. A day after assessing tumor appearance, tumors were ablated during diffusion-weighted imaging (DWI)-MRI using a Visualase LITT system (n = 5). Brain images were obtained immediately after ablation and again at 24 h post-ablation to confirm the efficacy of tumor cytoablation. Untreated tumors served as controls (n = 3). Rats were injected with fluorescent isothiocyanate (FITC) dextran and Evans blue that circulated for 10 min after post-LITT MRI. The brains were then removed for fluorescence microscopy and histopathology evaluations using hematoxylin and eosin (H&E) and major histocompatibility complex (MHC) staining. RESULTS All rats showed a space-occupying tumor with T2 and T1 contrast-enhancement at pre-LITT imaging. The rats that underwent the LITT procedure showed a well-demarcated ablation zone with near-complete ablation of tumor tissue and with peri-ablation contrast enhancement at 24 h post-ablation. Tumor cytoreduction by ablation as seen on MRI was confirmed by H&E and MHC staining. CONCLUSIONS Data showed that tumor cytoablation using MRI-monitored LITT was possible in preclinical glioma models. Real-time MRI monitoring facilitated visualizing and controlling the area of ablation as it is otherwise performed in clinical applications.
Collapse
|
21
|
Sadanandan N, Shear A, Brooks B, Saft M, Cabantan DAG, Kingsbury C, Zhang H, Anthony S, Wang ZJ, Salazar FE, Lezama Toledo AR, Rivera Monroy G, Vega Gonzales-Portillo J, Moscatello A, Lee JY, Borlongan CV. Treating Metastatic Brain Cancers With Stem Cells. Front Mol Neurosci 2021; 14:749716. [PMID: 34899179 PMCID: PMC8651876 DOI: 10.3389/fnmol.2021.749716] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Stem cell therapy may present an effective treatment for metastatic brain cancer and glioblastoma. Here we posit the critical role of a leaky blood-brain barrier (BBB) as a key element for the development of brain metastases, specifically melanoma. By reviewing the immunological and inflammatory responses associated with BBB damage secondary to tumoral activity, we identify the involvement of this pathological process in the growth and formation of metastatic brain cancers. Likewise, we evaluate the hypothesis of regenerating impaired endothelial cells of the BBB and alleviating the damaged neurovascular unit to attenuate brain metastasis, using the endothelial progenitor cell (EPC) phenotype of bone marrow-derived mesenchymal stem cells. Specifically, there is a need to evaluate the efficacy for stem cell therapy to repair disruptions in the BBB and reduce inflammation in the brain, thereby causing attenuation of metastatic brain cancers. To establish the viability of stem cell therapy for the prevention and treatment of metastatic brain tumors, it is crucial to demonstrate BBB repair through augmentation of vasculogenesis and angiogenesis. BBB disruption is strongly linked to metastatic melanoma, worsens neuroinflammation during metastasis, and negatively influences the prognosis of metastatic brain cancer. Using stem cell therapy to interrupt inflammation secondary to this leaky BBB represents a paradigm-shifting approach for brain cancer treatment. In this review article, we critically assess the advantages and disadvantages of using stem cell therapy for brain metastases and glioblastoma.
Collapse
Affiliation(s)
| | - Alex Shear
- University of Florida, Gainesville, FL, United States
| | - Beverly Brooks
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Madeline Saft
- University of Michigan, Ann Arbor, MI, United States
| | | | - Chase Kingsbury
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Henry Zhang
- University of Florida, Gainesville, FL, United States
| | - Stefan Anthony
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| | - Zhen-Jie Wang
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Felipe Esparza Salazar
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud (FCS), Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Alma R. Lezama Toledo
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud (FCS), Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Germán Rivera Monroy
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud (FCS), Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | | | - Alexa Moscatello
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| | - Cesario V. Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
- Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, United States
| |
Collapse
|
22
|
Cai Y, Chen T, Liu J, Peng S, Liu H, Lv M, Ding Z, Zhou Z, Li L, Zeng S, Xiao E. Orthotopic Versus Allotopic Implantation: Comparison of Radiological and Pathological Characteristics. J Magn Reson Imaging 2021; 55:1133-1140. [PMID: 34611963 PMCID: PMC9291575 DOI: 10.1002/jmri.27940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 11/11/2022] Open
Abstract
Background In experimental animal models, implantation location might influence the heterogeneity and overall development of the tumor, leading to an interpretation bias. Purpose To investigate the effects of implantation location in experimental tumor model using magnetic resonance imaging (MRI) and pathological findings. Study Type Prospective. Subjects Forty‐five breast cancer‐bearing mice underwent orthotopic (N = 15) and heterotopic (intrahepatic [N = 15] and subcutaneous [N = 15]) implantation. Field Strength/Sequence Sequences including: T1‐weighted turbo spin echo sequence, T2‐weighted blade sequence, diffusion‐weighted imaging, pre‐ and post‐contrast T1 mapping, multi‐echo T2 mapping at 3.0 T. Assessment MRI was performed at 7, 14, and 21 days after implantation. Native T1, post‐contrast T1, T2, and apparent diffusion coefficient (ADC) of tumors, the tumor volume and necrosis volume within tumor were obtained. Lymphocyte cells from H&E staining, Ki67‐positive, and CD31‐positive cells from immunohistochemistry were determined. Statistical Tests One‐way analysis of variance and Spearman's rank correlation were performed. P value <0.05 was considered statistically significant. Results The tumor volume (intrahepatic vs. orthotopic vs. subcutaneous: 587.50 ± 77.62 mm3 vs. 814.00 ± 43.85 mm3 vs. 956.13 ± 119.22 mm3), necrosis volume within tumor (89.10 ± 26.60 mm3 vs. 292.41 ± 57.92 mm3 vs. 179.91 ± 31.73 mm3, respectively), ADC at day 21 (543.41 ± 42.28 vs. 542.92 ± 99.67 vs. 369.83 ± 42.90, respectively), and post‐contrast T1 at all timepoints (day 7: 442.00 ± 11.52 vs. 435.00 ± 22.90 vs. 394.33 ± 29.95; day 14: 459.00 ± 26.11 vs. 436.83 ± 26.01 vs. 377.00 ± 27.83; day 21: 463.50 ± 23.49 vs. 458.00 ± 34.28 vs. 375.00 ± 30.55) were significantly different between three groups. Necrosis volumes of subcutaneous and intrahepatic tumors were significantly lower than those of orthotopic tumors. The CD31‐positive rate in the intrahepatic implantation was significantly higher than in orthotopic and subcutaneous groups. Necrosis volume (r = −0.71), ADC (r = −0.85), and post‐contrast T1 (r = −0.75) were strongly correlated with vascular invasion index. Data Conclusion Orthotopic and heterotopic tumors have their unique growth kinetics, necrosis volume, and vascular invasion. Non‐invasive MR quantitative parameters, including ADC and post‐contrast T1, may reflect vascular invasion in mice. Level of Evidence 1 Technical Efficacy Stage 3
Collapse
Affiliation(s)
- YeYu Cai
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - TaiLi Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - JiaYi Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - ShuHui Peng
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huan Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Min Lv
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - ZhuYuan Ding
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - ZiYi Zhou
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lan Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - EnHua Xiao
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China.,Molecular Imaging Research Center, Central South University, Changsha, China.,Clinical Research Center for Medical Imaging in Hunan Province, Changsha, China
| |
Collapse
|
23
|
Yang Z, Shi H, Chinnathambi A, Salmen SH, Alharbi SA, Veeraraghavan VP, Surapaneni KM, Arulselvan P. Arbutin exerts anticancer activity against rat C6 glioma cells by inducing apoptosis and inhibiting the inflammatory markers and P13/Akt/mTOR cascade. J Biochem Mol Toxicol 2021; 35:e22857. [PMID: 34338399 DOI: 10.1002/jbt.22857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/03/2021] [Accepted: 07/14/2021] [Indexed: 11/08/2022]
Abstract
Gliomas are a type of brain cancer that occurs in the supporting glial cells of the brain. It is highly malignant and accounts for 80% of brain tumors with high mortality and morbidity. Phytomedicines are potent alternatives for allopathic drugs which cause side effects. They have been used from ancient times by traditional Chinese, Ayurveda, and Siddha medicine. Arubtin is a glycoside phytochemical extracted from plants and belongs to the family of Ericaceae. Arbutin possesses various pharmacological properties such as anti-inflammatory, antioxidant, antitumor, and so on. Hence in the present study, we analyzed the anticancer potency of arbutin against rat C6 glioma cells. Rat C6 glioma cells were procured from American Type Culture Collection and the cells were cultured in Roswell Park Memorial Institute-1640 medium. To assess the cytotoxicity effect of the arbutin against C6 glioma cells, an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide test was performed with different doses from 10 to 60 µM. Arbutin effectively induced apoptosis in the cells and the IC50 dose was obtained at 30 µM. For further studies, we selected the 30 µM IC50 dose and a higher dose of 40 µM. Reactive oxygen species (ROS) generated were analyzed with DCFDA/H2DCFDA stain and the destruction of mitochondrial membrane permeability which is the initiator of apoptosis was analyzed with a cationic stain Rhodamine 123. Dual staining with acridine orange and ethidium bromide was performed to assess the viable and dead cells. Cell adhesion properties of glioma cells were analyzed with Matrigel assay. The apoptotic, inflammatory, and phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling molecules were analyzed with quantitative polymerase chain reaction (qPCR) analysis to confirm the anticancer effect of arbutin. Arbutin generated excessive ROS and disrupted the mitochondrial membrane, which induced apoptosis in cells, it also inhibited the cell adhesion property of C6 glioma cells. qPCR analysis clearly indicates arbutin increases the apoptotic genes and decreased the inflammatory and PI3K/mTOR signaling molecules. Overall, our results authentically confirm that arbutin can be a potent alternative for treating glioma.
Collapse
Affiliation(s)
- Zhangkai Yang
- Department of Neurosurgery, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Hangyu Shi
- Department of Neurosurgery, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Saleh H Salmen
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman A Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Krishna Mohan Surapaneni
- Department of Biochemistry, Clinical Skills & Simulation, Molecular Virology and Research, Panimalar Medical College Hospital & Research Institute, Chennai, Tamil Nadu, India
| | | |
Collapse
|
24
|
Deng YH, Yang YM, Ruan J, Mu L, Wang SQ. Effects of nursing care in fast-track surgery on postoperative pain, psychological state, and patient satisfaction with nursing for glioma. World J Clin Cases 2021; 9:5435-5441. [PMID: 34307597 PMCID: PMC8281405 DOI: 10.12998/wjcc.v9.i20.5435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/07/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The brain is the most complex organ in the human body. Treatment for a glioma always involves a multi-disciplinary team. Nursing care in fast-track surgery or enhanced recovery after surgery is such kind of work implemented by an interdisciplinary team to provide services to patients to improve their outcomes.
AIM To explore the effects of nursing care in fast-track surgery on postoperative pain, psychological state, and patient satisfaction with nursing for glioma.
METHODS From June 2018 to June 2020, 138 patients who underwent operation for glioma at Cancer Hospital Affiliated to Chongqing University were selected. They were categorized into groups according to different nursing care that they received. Of them, 69 patients receiving nursing care in fast-track surgery were included in an experimental group, and 69 patients receiving conventional postoperative nursing were included in a control group. Visual analogue scale was used to evaluate postoperative pain in the two groups immediately after the operation and at 3 d after the operation. Self-rating anxiety scale (SAS) and self-rating depression scale (SDS) were used to evaluate the psychological status of patients immediately after operation and on the 3rd postoperative day. A self-made satisfaction scale for patient satisfaction with nursing was used to evaluate and compare patient satisfaction with nursing between the two groups.
RESULTS Time to excretion, time to out-of-bed activities, and length of hospital stay were significantly shorter in the observation group than in the control group (P < 0.05). There was no significant difference in duration of operative time or intraoperative bleeding between the two groups (P > 0.05). There was no significant difference in postoperative pain score between the two groups (P > 0.05). The pain score was significantly lower in the observation group than in the control group at 3 d after the operation (P < 0.05). There was no significant difference in postoperative SAS or SDS score between the two groups (P > 0.05). SAS and SDS scores were significantly lower in the observation group than in the control group at 3 d after operation (P < 0.05). The rate of patient satisfaction with nursing was 94.2% in the observation group, which was significantly higher than that (81.2%) of the control group (P < 0.05).
CONCLUSION Nursing care in fast-track surgery can relieve postoperative pain, anxiety, and depression, and improve patient satisfaction with nursing in patients with glioma, which is worthy of clinical application.
Collapse
Affiliation(s)
- Yan-Hong Deng
- Department of Neurosurgery, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yi-Mei Yang
- Sterile Supply Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jian Ruan
- Department of Neurosurgery, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Lin Mu
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Shi-Qiang Wang
- Department of Neurosurgery, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
25
|
Hicks WH, Bird CE, Traylor JI, Shi DD, El Ahmadieh TY, Richardson TE, McBrayer SK, Abdullah KG. Contemporary Mouse Models in Glioma Research. Cells 2021; 10:cells10030712. [PMID: 33806933 PMCID: PMC8004772 DOI: 10.3390/cells10030712] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/20/2021] [Accepted: 03/20/2021] [Indexed: 02/07/2023] Open
Abstract
Despite advances in understanding of the molecular pathogenesis of glioma, outcomes remain dismal. Developing successful treatments for glioma requires faithful in vivo disease modeling and rigorous preclinical testing. Murine models, including xenograft, syngeneic, and genetically engineered models, are used to study glioma-genesis, identify methods of tumor progression, and test novel treatment strategies. Since the discovery of highly recurrent isocitrate dehydrogenase (IDH) mutations in lower-grade gliomas, there is increasing emphasis on effective modeling of IDH mutant brain tumors. Improvements in preclinical models that capture the phenotypic and molecular heterogeneity of gliomas are critical for the development of effective new therapies. Herein, we explore the current status, advancements, and challenges with contemporary murine glioma models.
Collapse
Affiliation(s)
- William H. Hicks
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
| | - Cylaina E. Bird
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
| | - Jeffrey I. Traylor
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
| | - Diana D. Shi
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA;
| | - Tarek Y. El Ahmadieh
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
| | - Timothy E. Richardson
- Department of Pathology, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX 75229, USA;
| | - Samuel K. McBrayer
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Harrold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Correspondence: (S.K.M.); (K.G.A.)
| | - Kalil G. Abdullah
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA; (W.H.H.); (C.E.B.); (J.I.T.); (T.Y.E.A.)
- Harrold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Correspondence: (S.K.M.); (K.G.A.)
| |
Collapse
|
26
|
Prashanth A, Donaghy H, Stoner SP, Hudson AL, Wheeler HR, Diakos CI, Howell VM, Grau GE, McKelvey KJ. Are In Vitro Human Blood-Brain-Tumor-Barriers Suitable Replacements for In Vivo Models of Brain Permeability for Novel Therapeutics? Cancers (Basel) 2021; 13:955. [PMID: 33668807 PMCID: PMC7956470 DOI: 10.3390/cancers13050955] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND High grade gliomas (HGG) are incapacitating and prematurely fatal diseases. To overcome the poor prognosis, novel therapies must overcome the selective and restricted permeability of the blood-brain barrier (BBB). This study critically evaluated whether in vitro human normal BBB and tumor BBB (BBTB) are suitable alternatives to "gold standard" in vivo models to determine brain permeability. METHODS A systematic review utilizing the PRISMA guidelines used English and full-text articles from the past 5 years in the PubMed, Embase, Medline and Scopus databases. Experimental studies employing human cell lines were included. RESULTS Of 1335 articles, the search identified 24 articles for evaluation after duplicates were removed. Eight in vitro and five in vivo models were identified with the advantages and disadvantages compared within and between models, and against patient clinical data where available. The greatest in vitro barrier integrity and stability, comparable to in vivo and clinical permeability data, were achieved in the presence of all cell types of the neurovascular unit: endothelial cells, astrocytes/glioma cells, pericytes and neurons. CONCLUSIONS In vitro co-culture BBB models utilizing stem cell-derived or primary cells are a suitable proxy for brain permeability studies in order to reduce animal use in medical research.
Collapse
Affiliation(s)
- Archana Prashanth
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Heather Donaghy
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Shihani P. Stoner
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Amanda L. Hudson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Helen R. Wheeler
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
- Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Connie I. Diakos
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
- Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Viive M. Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| | - Georges E. Grau
- Vascular Immunology, Department of Pathology, School of Pathology, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia;
| | - Kelly J. McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW 2065, Australia; (A.P.); (H.D.); (S.P.S.); (A.L.H.); (H.R.W.); (C.I.D.); (V.M.H.)
| |
Collapse
|
27
|
WANG KAI, ZHANG FENGTIAN, WEN CHANGLONG, HUANG ZHIHUA, HU ZHIHAO, ZHANG YUWEN, HU FUQIANG, WEN LIJUAN. Regulation of pathological blood-brain barrier for intracranial enhanced drug delivery and anti-glioblastoma therapeutics. Oncol Res 2021. [DOI: 10.32604/or.2022.025696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
|
28
|
Ma YS, Yang XL, Xin R, Liu JB, Fu D. Power and promise of exosomes as clinical biomarkers and therapeutic vectors for liquid biopsy and cancer control. Biochim Biophys Acta Rev Cancer 2020; 1875:188497. [PMID: 33370570 DOI: 10.1016/j.bbcan.2020.188497] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
Exosomes, microvesicles derived from the nuclear endosome and plasma membrane, can be released into the extracellular environment to act as mediators between the cell membrane and cytoplasmic proteins, lipids, or RNA. Exosomes are considered effective carriers of intercellular signals in prokaryotes and eukaryotes, because of their ability to efficiently transfer proteins, lipids, and nucleic acids between cellular compartments. Since the 2007 discovery that exosomes carry bioactive substances, exosomes have been intensively researched. In various physiological and pathological processes, exosomes play important biological roles by specifically combining with receptor cells and transmitting information. Their stable biological characteristics, diversity of contents, non-invasiveness path for introducing signaling molecules, and ability for rapid detection make exosomes a promising clinical diagnostic marker for potentially many pathological conditions, including cancers. Exosomes are not only considered biomarkers and prognostic disease factors, but also have potential as gene carriers and drug delivery vectors, and have important clinical significance and application potential in the fields of cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Yu-Shui Ma
- Department of Pancreatic and Hepatobiliary Surgery, Cancer Hospital, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Cancer Institute, Nantong Tumor Hospital, Nantong 226631, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| | - Xiao-Li Yang
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Rui Xin
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ji-Bin Liu
- Cancer Institute, Nantong Tumor Hospital, Nantong 226631, China
| | - Da Fu
- Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|