1
|
Kim YB, Lee SH, Kasala D, Zhao Y, Jiao A, Hong J, Kim JS, Yoon AR, Yun CO. Potent therapeutic efficacy of intranasally deliverable paclitaxel modified with pH-sensitive and PEGylated polymeric micelle against glioblastoma. J Control Release 2025; 382:113711. [PMID: 40204132 DOI: 10.1016/j.jconrel.2025.113711] [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: 11/08/2024] [Revised: 03/10/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and common type of brain tumor. Conventional therapies for GBM, such as surgery or radiotherapy, have shown inadequate therapeutic effect. Similarly, a large fraction of chemotherapeutics are ineffective against GBM due to the blood-brain barrier (BBB) preventing effective delivery of these drugs to the brain. To overcome these obstacles, an intranasally administrable and multifunctional drug-loaded polymeric micelle composed of a pH-sensitive PPCBA-PEI-Arg (PPA) polymer conjugated with PEGylated paclitaxel (PEG-PTX; PPP) was synthesized to treat GBM. PPP was more soluble in an aqueous solution than parental PTX and was more effectively internalized into the GBM cells. Further, PPP elicited a more potent cancer cell killing effect than PTX under physiological pH condition, which was further augmented under the mildly acidic condition that emulated the tumor microenvironment. Intranasal administration of PPP into orthotopic GBM tumor xenograft-bearing mice led to more efficient delivery of the drug to the brain tissues compared to parental PTX delivered via intranasal or intravenous route, thus resulting in superior inhibition of GBM growth. Collectively, these findings demonstrated that intranasal delivery of PTX via pH-sensitive and PEGylated polymeric micelles can be an effective approach for the treatment of aggressive GBM.
Collapse
Affiliation(s)
- Young-Beom Kim
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - Soo-Hwan Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - Dayananda Kasala
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - Yuebin Zhao
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - Ao Jiao
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - JinWoo Hong
- GeneMedicine Co., Ltd., Seoul, Republic of Korea
| | - Jin Su Kim
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul, Republic of Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, Republic of Korea; Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, Republic of Korea; Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea; GeneMedicine Co., Ltd., Seoul, Republic of Korea.
| |
Collapse
|
2
|
Pathak D, Singh KP. Neurobehavioral manifestations in female rats after intermittent exposure to an anticancer agent, paclitaxel. Behav Pharmacol 2025:00008877-990000000-00135. [PMID: 40397101 DOI: 10.1097/fbp.0000000000000833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Paclitaxel (PTX), a widely used chemotherapeutic agent, causes both peripheral and central neurotoxicity, leading to significant behavioral impairments. However, inadequate literature is available on PTX-induced neurobehavioral sequelae associated with anxiety, depression, and cognition in adults during and after chemotherapy. Therefore, the present study aimed to investigate neurobehavioral impairments in adult female rats following PTX exposure, with a specific focus on anxiety-like behaviors and cognitive functions such as learning and memory. In this study, we used adult female Wistar rats aged 10-12 weeks (average weight: 180 ± 5 g) and administered clinically relevant therapeutic doses of PTX (1.6 and 3.2 mg/kg body weight) intravenously once weekly for 6 weeks, simulating the clinical chemotherapy regimen. Neurobehavioral assessments were conducted after the first and sixth doses of PTX using validated mazes, including the photoactometer, open-field maze, elevated plus-maze (EPM; for anxiety-like behaviors), and the step-down latency test (SDL; for cognitive performance). Neurobehavioral patterns were recorded using autotracking software (ANY-maze, Stoelting Co., Wood Dale, Illinois, USA). Our findings revealed substantially reduced locomotor activity in the photoactometer, increased anxiety-like behaviors with amplified fear emotionality in the open-field and EPM tests, and memory impairment in the SDL test. These results suggest that the manifestation of anxiogenic and cognitive behavioral changes is associated with the administration of a higher dose (3.2 mg/kg) of PTX. In conclusion, our study indicates that PTX causes significant neurobehavioral impairments in rats after exposure to equivalent therapeutic doses of PTX.
Collapse
Affiliation(s)
- Deepika Pathak
- Neurobiology Laboratory, Department of Zoology, University of Allahabad, Prayagraj, India
| | | |
Collapse
|
3
|
Shi H, Wang B, Shi Z, Ma H, Li Y, Liu Y, Zhao Y, Xia N, Wu C, Gao Y. Paclitaxel-Ang-2-functionalized bionic mesoporous selenium nanoparticles for targeted therapy of glioma. Pharmacol Res 2025; 216:107783. [PMID: 40378941 DOI: 10.1016/j.phrs.2025.107783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/19/2025]
Abstract
Glioma, the most prevalent primary intracranial tumor, presents significant clinical treatment challenges due to its high invasiveness and therapeutic resistance. Therefore, the development of a targeted therapeutic agent that is both highly effective and low in toxicity is crucial. In this research, we aimed to design a bionic mesoporous selenium nanoparticle (ACMLMSeP) functionalized with paclitaxel and Ang-2 for nasal administration as a targeted treatment approach for glioma. Nasal administration facilitates direct delivery of drugs to the brain through the olfactory nerve, thereby circumventing the protective mechanisms of the blood-brain barrier. Mesoporous selenium (MSe) significantly enhances the loading capacity for insoluble drugs while improving their water solubility. The functionalization of MSe enables slow drug release and facilitates targeted drug accumulation. Moreover, accumulated nano-selenium promotes reactive oxygen species (ROS) production, induces autophagy, and synergizes with drugs to accelerate apoptosis in tumor cells. Analysis using Transmission Electron Microscopy (TEM) images and Dynamic Light Scattering (DLS) indicated that ACMLMSe has an average particle size of roughly 135 nm. Results from in vitro release assessments indicated that the ACMLMSeP sustained the release of the drug, reaching a total release rate of 74.96 ± 2.34 % within 24 h. Cellular uptake studies and in vivo imaging showed the strong targeting capabilities of the ACMLMSeP nanoparticles. Furthermore, the results obtained from the MTT assays, flow cytometry analysis, immunofluorescence staining, and in vivo antitumor evaluations collectively revealed that ACMLMSeP effectively inhibited proliferation while promoting apoptosis in C6 cells. In summary, these experimental findings clearly suggest that ACMLMSeP may serve as a promising biomimetic nanosystem for the targeted treatment of brain glioma.
Collapse
Affiliation(s)
- Huan Shi
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Biaobiao Wang
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Zhiwei Shi
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Huilin Ma
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Yunmei Li
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Ying Liu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Yu Zhao
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Nan Xia
- Xuzhou Central Hospital, Quanshan District, Xuzhou, Jiangsu, China.
| | - Chao Wu
- Pharmacy School, Jinzhou Medical University, 40 Songpo Road, Linghe, Jinzhou, Liaoning 121001, China.
| | - Yu Gao
- Department of Medical Oncology, the First Affiliated Hospital of Jinzhou Medical University, No.2, the Fifth Section of Renmin Street, Guta District, Jinzhou, Liaoning 121001, China.
| |
Collapse
|
4
|
Shokri Varniab Z, Chang E, Wang J, Duwa R, Suryadevara V, Wu W, Kumar M, Liang T, Khatoon Z, Morais GR, Falconer R, Shi Y, Tikhomirov G, Nernekli K, Pisani LJ, Daldrup-Link HE. Dual-enzyme activated theranostic nanoparticles for image-guided glioblastoma therapy. Sci Rep 2025; 15:13540. [PMID: 40253484 PMCID: PMC12009400 DOI: 10.1038/s41598-025-97775-w] [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: 01/31/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025] Open
Abstract
Matrix metalloproteinase-14 (MMP-14) and Cathepsin-B (Cat-B) are overexpressed in glioblastoma (GBM) and not normal brain, making them promising targets for prodrug activation. We investigated a novel combination therapy using two tumor-enzyme activatable theranostic nanoprobes (TNP): TNP-MMP-14, which disrupts the blood tumor barrier via MMP-14 activation, and TNP-Cat-B, which selectively targets GBM cells through Cat-B activation. We hypothesized that combining TNP-MMP-14 and TNP-Cat-B would enhance TNP tumor accumulation and therapeutic efficacy compared to TNP-Cat-B monotherapy. Thirty NSG mice with luciferase-expressing GBM39 tumors received either TNP-MMP-14 plus TNP-Cat-B, TNP-Cat-B only, or saline. Magnetic resonance imaging (MRI) was conducted pre- and post-treatment, with T2* relaxation times analyzed using a generalized linear model. Histopathological differences were assessed using Kruskal-Wallis and Mann-Whitney tests. A Bonferroni correction was applied to account for multiple comparisons. Combination therapy significantly reduced tumor T2* relaxation times (12.98 ± 4.20 ms) compared to TNP-Cat-B monotherapy (22.49 ± 3.95 ms, p < 0.001). The apoptotic marker caspase-3 was also significantly higher in the combination group (64.46 ± 23.43 vs. 15.93 ± 5.81, p < 0.001). These findings demonstrate the potential of dual-enzyme activatable nanoparticles to enhance GBM treatment by overcoming drug delivery barriers and improving therapeutic efficacy over monotherapy.
Collapse
Affiliation(s)
- Zahra Shokri Varniab
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
| | - Edwin Chang
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
- Stanford Center for Innovation in In Vivo Imaging (SCi3) at Porter, Canary Center for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Jie Wang
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
| | - Ramesh Duwa
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
| | - Vidyani Suryadevara
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
| | - Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
| | - Manoj Kumar
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
| | - Tie Liang
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
| | - Zubeda Khatoon
- Faculty of Life Sciences, Institute of Cancer Therapeutics, University of Bradford, Bradford, UK
| | - Goreti Ribeiro Morais
- Faculty of Life Sciences, Institute of Cancer Therapeutics, University of Bradford, Bradford, UK
| | - Robert Falconer
- Faculty of Life Sciences, Institute of Cancer Therapeutics, University of Bradford, Bradford, UK
| | - Yifeng Shi
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
| | - Grigory Tikhomirov
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
| | - Kerem Nernekli
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
| | - Laura Jean Pisani
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA
| | - Heike Elisabeth Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, 725 Welch Rd, Stanford, CA, 94305-5614, USA.
- Department of Pediatrics, Pediatric Oncology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
5
|
Jiang X, Lai Y, Xia W, Yang W, Wang J, Pan J, Zhao Q, Zhou F, Li S, Zhang S, Gao J, Wang Y, Zan T, Xu ZP, Yu H, Xu Z. Self-Oxygenating PROTAC Microneedle for Spatiotemporally-Confined Protein Degradation and Enhanced Glioblastoma Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411869. [PMID: 40025927 DOI: 10.1002/adma.202411869] [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: 08/11/2024] [Revised: 12/31/2024] [Indexed: 03/04/2025]
Abstract
Glioblastoma (GBM) is the most aggressive subtype of primary brain tumors, which marginally respond to standard chemotherapy due to the blood-brain barrier (BBB) and the low tumor specificity of the therapeutics. Herein, a double-layered microneedle (MN) patch is rationally engineered by integrating acid and light dual-activatable PROteolysis TArgeting Chimera (PROTAC) nanoparticles and self-oxygenating BSA-MnO2 (BM) nanoparticles for GBM treatment. The MN is administrated at the tumor site to locally deliver the PROTAC prodrug and BM nanoparticles. The PROTAC nanoparticles are rapidly released from the outer layer of the MN and specifically activated in the acidic intracellular environment of tumor cells. Subsequently, near-infrared light activates the photosensitizer to produce singlet oxygen (1O2) through photodynamic therapy (PDT), thereby triggering spatiotemporally-tunable degradation of bromodomain and extraterminal protein 4 (BRD4). The BM nanoparticles, in the inner layer of the MN, serve as an oxygen supply station, and counteracts tumor hypoxia by converting hydrogen peroxide (H2O2) into oxygen (O2), thus promoting PDT and PROTAC activation. This PROTAC prodrug-integrated MN significantly inhibits tumor growth in both subcutaneous and orthotopic GBM tumor models. This study describes the first spatiotemporally-tunable protein degradation strategy for highly efficient GBM therapy, potentially advancing precise therapy of other kinds of refractory brain tumors.
Collapse
Affiliation(s)
- Xingyu Jiang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yi Lai
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wenzheng Xia
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Wenfang Yang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Junjue Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiaxing Pan
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qian Zhao
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Feng Zhou
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shiqin Li
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shunan Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Jing Gao
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhi Ping Xu
- Institute of Biomedical Health Technology and Engineering and Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518067, China
| | - Haijun Yu
- State Key Laboratory of Chemical Biology and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| |
Collapse
|
6
|
Géraudie A, De Rossi P, Canney M, Carpentier A, Delatour B. Effects of blood-brain barrier opening using ultrasound on tauopathies: A systematic review. J Control Release 2025; 379:1029-1044. [PMID: 39875073 DOI: 10.1016/j.jconrel.2025.01.056] [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: 11/22/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
Blood-brain barrier opening with ultrasound can potentiate drug efficacy in the treatment of brain pathologies and also provides therapeutic effects on its own. It is an innovative tool to transiently, repeatedly and safely open the barrier, with studies showing beneficial effects in both preclinical models for Alzheimer's disease and recent clinical studies. The first preclinical and clinical work has mainly shown a decrease in amyloid burden in mice models and in patients. However, Alzheimer's disease pathology also encompasses tauopathy, which is closely related to cognitive decline, making it a crucial therapeutic target. The effects of blood-brain barrier opening with ultrasound have been rarely assessed on tau and are still unclear. METHODS This systematic review, conducted through searches using Pubmed, Embase, Web of Science and Cochrane Central databases, extracted results of 15 studies reporting effects of blood-brain barrier opening using ultrasound on tau proteins. RESULTS This review of the literature indicates that blood-brain barrier opening using ultrasound can decrease the extent of the tau pathology or potentialize the effect of a therapeutic drug. However, selected studies report paradoxically that blood-brain barrier opening can increase tau pathology burden and induce brain damage. DISCUSSION Apparent discrepancies between reports could originate from the variability in protocols or analytical methods that may impact the effects of blood-brain barrier opening with ultrasound on tauopathies, glial populations, tissue integrity and functional outcomes. CONCLUSION This calls for a better standardization effort combined with improved methodologies allowing between-studies comparisons, and for further understanding of the effects of blood-brain barrier opening on tau pathology as an essential prerequisite before translation to clinic.
Collapse
Affiliation(s)
- Amandine Géraudie
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France.
| | | | | | - Alexandre Carpentier
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France; Faculty of Medicine, Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France; Advanced Surgical Research Technology Lab, Sorbonne University, 75013 Paris, France
| | - Benoît Delatour
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France
| |
Collapse
|
7
|
Pedder JH, Sonabend AM, Cearns MD, Michael BD, Zakaria R, Heimberger AB, Jenkinson MD, Dickens D. Crossing the blood-brain barrier: emerging therapeutic strategies for neurological disease. Lancet Neurol 2025; 24:246-260. [PMID: 39862873 DOI: 10.1016/s1474-4422(24)00476-9] [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: 03/28/2024] [Revised: 09/20/2024] [Accepted: 11/12/2024] [Indexed: 01/27/2025]
Abstract
The blood-brain barrier is a physiological barrier that can prevent both small and complex drugs from reaching the brain to exert a pharmacological effect. For treatment of neurological diseases, drug concentrations at the target site are a fundamental parameter for therapeutic effect; thus, the blood-brain barrier is a major obstacle to overcome. Novel strategies have been developed to circumvent the blood-brain barrier, including CSF delivery, intracranial delivery, ultrasound-based methods, membrane transporters, receptor-mediated transcytosis, and nanotherapeutics. These approaches each have their advantages and disadvantages. CSF delivery and intracranial delivery are direct but invasive techniques that have not yet shown efficacy in clinical trials, although development of novel delivery devices might improve these approaches. Ultrasound-based disruption has shown some efficacy in clinical trials, but it can require invasive procedures. Approaches using membrane transporters and receptor-mediated transcytosis are less invasive than are other techniques, but they can have off-target effects. Nanotherapeutics have shown promise, but these strategies are in early stages of development. Advancements in drug delivery across the blood-brain barrier will require appropriately designed and powered clinical studies, with a focus on the timing of treatment, demographic and genetic considerations, head-to-head comparison with other treatment strategies (rather than a placebo), and relevant primary and secondary outcome measures.
Collapse
Affiliation(s)
- Josephine H Pedder
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Adam M Sonabend
- Department of Neurological Surgery, Malnati Brain Tumor Institute of the Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael D Cearns
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Benedict D Michael
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Rasheed Zakaria
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - Amy B Heimberger
- Department of Neurological Surgery, Malnati Brain Tumor Institute of the Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Michael D Jenkinson
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK; Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - David Dickens
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
8
|
Sun B, Li R, Ji N, Liu H, Wang H, Chen C, Bai L, Su J, Chen J. Brain-targeting drug delivery systems: The state of the art in treatment of glioblastoma. Mater Today Bio 2025; 30:101443. [PMID: 39866779 PMCID: PMC11759563 DOI: 10.1016/j.mtbio.2025.101443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Glioblastoma (GBM) is the most prevalent primary malignant brain tumor, characterized by a high mortality rate and a poor prognosis. The blood-brain barrier (BBB) and the blood-tumor barrier (BTB) present significant obstacles to the efficacy of tumor-targeted pharmacotherapy, thereby impeding the therapeutic potential of numerous candidate drugs. Targeting delivery of adequate doses of drug across the BBB to treat GBM has become a prominent research area in recent years. This emphasis has driven the exploration and evaluation of diverse technologies for GBM pharmacotherapy, with some already undergoing clinical trials. This review provides a thorough overview of recent advancements and challenges in targeted drug delivery for GBM treatment. It specifically emphasizes systemic drug administration strategies to assess their potential and limitations in GBM treatment. Furthermore, this review highlights promising future research directions in the development of intelligent drug delivery systems aimed at overcoming current challenges and enhancing therapeutic efficacy against GBM. These advancements not only support foundational research on targeted drug delivery systems for GBM but also offer methodological approaches for future clinical applications.
Collapse
Affiliation(s)
- Bo Sun
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Rong Li
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ning Ji
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Hongxiang Wang
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chao Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Trauma Orthopedics Center, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
9
|
Zhang G, Xu Y, Zhou A, Yu Y, Ning X, Bao H. Bioengineered NanoAid synergistically targets inflammatory pro-tumor processes to advance glioblastoma chemotherapy. NANOSCALE 2025; 17:2753-2768. [PMID: 39831463 DOI: 10.1039/d4nr04557b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Through transcriptomic analysis of patient-derived glioblastoma tissues, we identify an overactivation of inflammatory pathways that contribute to the development of a tumor-promoting microenvironment and therapeutic resistance. To address this critical mechanism, we present NanoAid, a biomimetic nanoplatform designed to target inflammatory pro-tumor processes to advance glioblastoma chemotherapy. NanoAid employs macrophage-membrane-liposome hybrids to optimize the delivery of COX-2 inhibitor parecoxib and paclitaxel. By inheriting macrophage characteristics, NanoAid not only efficiently traverses the blood-brain barrier and precisely accumulates within tumors but also enhances cancer cell uptake, thereby improving overall anticancer efficacy. Notably, the combination of parecoxib and paclitaxel effectively disrupts inflammatory pro-tumor processes while inducing a synergistic effect that inhibits tumor growth, overcomes therapeutic resistance, and minimizes adverse effects. This results in substantial tumor growth inhibition and extends the median survival of tumor-bearing mice. Thus, our study bridges clinical insights with fundamental research, potentially revolutionizing tumor therapy paradigms.
Collapse
Affiliation(s)
- Gui Zhang
- The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Yongle Yu
- Medical College of Guangxi University, Nanning 530004, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Hongguang Bao
- Department of Anaesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 211101, China.
| |
Collapse
|
10
|
Mundžić M, Ultimo A, Mladenović M, Pavlović A, Gobbo OL, Ruiz-Hernandez E, Santos-Martinez MJ, Knežević NŽ. Chlorotoxin-functionalized mesoporous silica nanoparticles for pH-responsive paclitaxel delivery to Glioblastoma multiforme. Heliyon 2025; 11:e41151. [PMID: 39758413 PMCID: PMC11699378 DOI: 10.1016/j.heliyon.2024.e41151] [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: 10/17/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain cancer associated with poor survival rates. We developed novel mesoporous silica nanoparticles (MSNs)-based nanocarriers for pH-responsive delivery of a therapeutic drug Paclitaxel (PTX) to GBM tumor cells. The pores of MSNs are loaded with PTX, which is retained by β-cyclodextrin (CD) moieties covalently linked to the pore entrances through a hydrazone linkage, which is cleavable in weakly acidic environment. Furthermore, we utilized a host-guest interaction between the adamantane and capping CD moieties to further functionalize the surface with a potential glioma-targeting oligopeptide chlorotoxin (CHX). In vitro studies in the U87 GBM cell line show decreased uptake, but increased toxicity of CHX-modified nanoparticles compared to CHX-free nanoparticles. The obtained results are promising toward development of advanced drug nanocarriers, which may target the overexpressed receptors in cancer tissues and utilize their weakly acidic environment for triggering the drug release, potentially leading to more efficient cancer treatments.
Collapse
Affiliation(s)
- Mirjana Mundžić
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000, Novi Sad, Serbia
| | - Amelia Ultimo
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02PN40, Dublin, Ireland
| | - Minja Mladenović
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000, Novi Sad, Serbia
| | - Aleksandra Pavlović
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02PN40, Dublin, Ireland
| | - Oliviero L. Gobbo
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02PN40, Dublin, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, D08 NHY1, Dublin, Ireland
| | - Eduardo Ruiz-Hernandez
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02PN40, Dublin, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, D08 NHY1, Dublin, Ireland
| | - Maria Jose Santos-Martinez
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02PN40, Dublin, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, D08 NHY1, Dublin, Ireland
- School of Medicine, Trinity College Dublin, D02 E8C0, Dublin, Ireland
| | - Nikola Ž. Knežević
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000, Novi Sad, Serbia
| |
Collapse
|
11
|
Gould A, Luan Y, Hou Y, Korobova FV, Chen L, Arrieta VA, Amidei C, Ward R, Gomez C, Castro B, Habashy K, Zhang D, Youngblood M, Dmello C, Bebawy J, Bouchoux G, Stupp R, Canney M, Yue F, Iruela-Arispe ML, Sonabend AM. Endothelial response to blood-brain barrier disruption in the human brain. JCI Insight 2024; 10:e187328. [PMID: 39724015 PMCID: PMC11949064 DOI: 10.1172/jci.insight.187328] [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] [Indexed: 12/28/2024] Open
Abstract
Cerebral endothelial cell (EC) injury and blood-brain barrier (BBB) permeability contribute to neuronal injury in acute neurological disease states. Preclinical experiments have used animal models to study this phenomenon, yet the response of human cerebral ECs to BBB disruption remains unclear. In our phase I clinical trial (ClinicalTrials.gov NCT04528680), we used low-intensity pulsed ultrasound with microbubbles (LIPU/MB) to induce transient BBB disruption of peritumoral brain in patients with recurrent glioblastoma. We found radiographic evidence that BBB integrity was mostly restored within 1 hour of this procedure. Using single-cell RNA sequencing and transmission electron microscopy, we analyzed the acute response of human brain ECs to ultrasound-mediated BBB disruption. Our analysis revealed distinct EC gene expression changes after LIPU/MB, particularly in genes related to neurovascular barrier function and structure, including changes to genes involved in the basement membrane, EC cytoskeleton, and junction complexes, as well as caveolar transcytosis and various solute transporters. Ultrastructural analysis showed that LIPU/MB led to a decrease in luminal caveolae, the emergence of cytoplasmic vacuoles, and the disruption of the basement membrane and tight junctions, among other things. These findings suggested that acute BBB disruption by LIPU/MB led to specific transcriptional and ultrastructural changes and could represent a conserved mechanism of BBB repair after neurovascular injury in humans.
Collapse
Affiliation(s)
- Andrew Gould
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| | - Yu Luan
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ye Hou
- Institute of Biomedicine, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Farida V. Korobova
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Li Chen
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| | - Victor A. Arrieta
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| | - Christina Amidei
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| | - Rachel Ward
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| | - Cristal Gomez
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| | - Brandyn Castro
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
- Department of Neurosurgery, University of Chicago, Chicago, Illinois, USA
| | - Karl Habashy
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| | - Daniel Zhang
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
- Rush Medical College, Chicago, Illinois, USA
| | - Mark Youngblood
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| | - Crismita Dmello
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| | - John Bebawy
- Department of Neurological Surgery
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Roger Stupp
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
- Department of Neurology and
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Feng Yue
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - M. Luisa Iruela-Arispe
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, and
| |
Collapse
|
12
|
Ćwiklińska A, Przewodowska D, Koziorowski D, Szlufik S. Innovative Approaches to Brain Cancer: The Use of Magnetic Resonance-guided Focused Ultrasound in Glioma Therapy. Cancers (Basel) 2024; 16:4235. [PMID: 39766134 PMCID: PMC11674718 DOI: 10.3390/cancers16244235] [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: 11/11/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Gliomas are a wide group of common brain tumors, with the most aggressive type being glioblastoma multiforme (GBM), with a 5-year survival rate of less than 5% and a median survival time of approximately 12-14 months. The standard treatment of GBM includes surgical excision, radiotherapy, and chemotherapy with temozolomide (TMZ). However, tumor recurrence and progression are common. Therefore, more effective treatment for GBM should be found. One of the main obstacles to the treatment of GBM and other gliomas is the blood-brain barrier (BBB), which impedes the penetration of antitumor chemotherapeutic agents into glioblastoma cells. Nowadays, one of the most promising novel methods for glioma treatment is Magnetic Resonance-guided Focused Ultrasound (MRgFUS). Low-intensity FUS causes the BBB to open transiently, which allows better drug delivery to the brain tissue. Under magnetic resonance guidance, ultrasound waves can be precisely directed to the tumor area to prevent side effects in healthy tissues. Through the open BBB, we can deliver targeted chemotherapeutics, anti-tumor agents, immunotherapy, and gene therapy directly to gliomas. Other strategies for MRgFUS include radiosensitization, sonodynamic therapy, histotripsy, and thermal ablation. FUS can also be used to monitor the treatment and progression of gliomas using blood-based liquid biopsy. All these methods are still under preclinical or clinical trials and are described in this review to summarize current knowledge and ongoing trials.
Collapse
Affiliation(s)
| | | | | | - Stanisław Szlufik
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 03-242 Warsaw, Poland
| |
Collapse
|
13
|
Pourmasoumi P, Banihashemian SA, Zamani F, Rasouli-Nia A, Mehrabani D, Karimi-Busheri F. Nanoparticle-Based Approaches in the Diagnosis and Treatment of Brain Tumors. J Clin Med 2024; 13:7449. [PMID: 39685907 DOI: 10.3390/jcm13237449] [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: 09/20/2024] [Revised: 11/28/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Glioblastomas are highly invasive brain tumors among perilous diseases. They are characterized by their fast proliferation and delayed detection that render them a significant focal point for medical research endeavors within the realm of cancer. Among glioblastomas, Glioblastoma multiforme (GBM) is the most aggressive and prevalent malignant brain tumor. For this, nanomaterials such as metallic and lipid nanoparticles and quantum dots have been acknowledged as efficient carriers. These nano-materials traverse the blood-brain barrier (BBB) and integrate and reach the necessary regions for neuro-oncology imaging and treatment purposes. This paper provides a thorough analysis on nanoparticles used in the diagnosis and treatment of brain tumors, especially for GBM.
Collapse
Affiliation(s)
- Parvin Pourmasoumi
- Department of Biomedical Engineering, Central Tehran Branch, Islamic Azad University, Tehran 19395-1495, Iran
- Stem Cells Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran 14778-93780, Iran
| | - Seyed Abdolvahab Banihashemian
- Department of Biomedical Engineering, Central Tehran Branch, Islamic Azad University, Tehran 19395-1495, Iran
- Stem Cells Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran 14778-93780, Iran
| | - Farshid Zamani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-69411, Iran
| | - Aghdass Rasouli-Nia
- Department of Oncology, Faculty of Medicine, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Davood Mehrabani
- Department of Oncology, Faculty of Medicine, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Comparative and Experimental Medicine Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Feridoun Karimi-Busheri
- Department of Oncology, Faculty of Medicine, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
14
|
Zhu S, Wu Q, Ying Y, Mao Y, Lu W, Xu J, Cai X, He H, Wu J. Tissue-Adaptive BSA Hydrogel with Dual Release of PTX and bFGF Promotes Spinal Cord Injury Repair via Glial Scar Inhibition and Axon Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401407. [PMID: 39385643 DOI: 10.1002/smll.202401407] [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: 02/22/2024] [Revised: 09/18/2024] [Indexed: 10/12/2024]
Abstract
Spinal cord injury (SCI) is a severe clinical disease usually accompanied by activated glial scar, neuronal axon rupture, and disabled motor function. To mimic the microenvironment of the SCI injury site, a hydrogel system with a comparable mechanical property to the spinal cord is desirable. Therefore, a novel elastic bovine serum albumin (BSA) hydrogel is fabricated with excellent adhesive, injectable, and biocompatible properties. The hydrogel is used to deliver paclitaxel (PTX) together with basic fibroblast growth factor (bFGF) to inhibit glial scar formation as well as promote axon regeneration and motor function for SCI repair. Due to the specific interaction of BSA with both drugs, bFGF, and PTX can be controllably released from the hydrogel system to achieve an effective concentration at the wound site during the SCI regeneration process. Moreover, benefiting from the combination of PTX and bFGF, this bFGF/PTX@BSA system significantly aided axon repair by promoting the elongation of axons across the glial scar with reduced reactive astrocyte secretion. In addition, remarkable anti-apoptosis of nerve cells is evident with the bFGF/PTX@BSA system. Subsequently, this multi-functionalized drug system significantly improved the motor function of the rats after SCI. These results reveal that bFGF/PTX@BSA is an ideal functionalized material for nerve repair in SCI.
Collapse
Affiliation(s)
- Sipin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Qiuji Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yibo Ying
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yuqin Mao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenjie Lu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jie Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiong Cai
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Huacheng He
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Jiang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
15
|
Ao H, Fu Y, Wang X. A comparative study of PEO-PBO content on the targeting and anti-glioma activity of annonaceous acetogenins-loaded nanomicelles. Colloids Surf B Biointerfaces 2024; 244:114176. [PMID: 39217726 DOI: 10.1016/j.colsurfb.2024.114176] [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: 06/27/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Annonaceous acetogenins (ACGs) have great potential in the treatment of gliomas, but are extremely insoluble and difficult for delivery in vivo. Poly(ethylene oxide)-b-poly(butylene oxide) (PEO-PBO) is an amphiphilic polymer and can reduce the clearance of nanoparticles by mononuclear phagocyte system. To explore an efficient and safe nanomedicine for glioma, ACGs-loaded nanomicelles (ACGs/EB-NCs) was constructed using PEO-PBO as a carrier, and the effect of PEO-PBO content on the targeting and anti-glioma activity were also compared. ACGs/EB5-NCs, ACGs/EB10-NCs and ACGs/EB20-NCs, the three nanomicellels prepared with different ACGs/EB feeding ratios, had average particle sizes of 148.8±0.5 nm, 32.7±4.1 nm, and 27.1±0.3 nm, respectively. The three ACGs/EB-NCs were spherical in shape, with drug loading content close to the theoretical drug loading content, encapsulation efficiency greater than 97 %, and good stability in physiological media. The cumulative release rates of ACGs/EB5-NCs, ACGs/EB10-NCs and ACGs/EB20-NCs were 78.2 %, 63.4 %, and 56.3 % within 216 hours, respectively. The inhibitory effects of three ACGs/EB-NCs on U87 MG cells were similar and stronger than free ACGs (P<0.05), with half inhibitory concentration of 0.17, 0.18, and 0.16 ng/mL (P>0.05), respectively. In U87 MG tumor‑bearing mice, ACGs/EB5-NC, ACGs/EB10-NCs and ACGs/EB20-NCs showed a similar tumor inhibition rate of 61.1±5.9 %, 56.2±8.6 % and 64.3±9.4 % (P>0.05), with good safety. Three ACGs/EB-NCs exhibited excellent liver escape ability and tumor targeting ability, with the tumor targeting index greater than 1.5. Three ACGs/EB-NCs were successfully prepared with strong anti-glioma activity and tumor targeting properties, which are expected to provide new options for the clinical treatment of gliomas. The content of PEO-PBO in micelles did not have a significant effect on the tumor targeting and anti-glioma activity of ACGs/EB-NCs.
Collapse
Affiliation(s)
- Hui Ao
- Department of Pharmacy, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi 214023, PR China
| | - Yao Fu
- Department of Pharmacy, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, No. 299, Qingyang Road, Liangxi District, Wuxi 214023, PR China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing 100193, PR China.
| |
Collapse
|
16
|
Kim KS, Habashy K, Gould A, Zhao J, Najem H, Amidei C, Saganty R, Arrieta VA, Dmello C, Chen L, Zhang DY, Castro B, Billingham L, Levey D, Huber O, Marques M, Savitsky DA, Morin BM, Muzzio M, Canney M, Horbinski C, Zhang P, Miska J, Padney S, Zhang B, Rabadan R, Phillips JJ, Butowski N, Heimberger AB, Hu J, Stupp R, Chand D, Lee-Chang C, Sonabend AM. Fc-enhanced anti-CTLA-4, anti-PD-1, doxorubicin, and ultrasound-mediated blood-brain barrier opening: A novel combinatorial immunotherapy regimen for gliomas. Neuro Oncol 2024; 26:2044-2060. [PMID: 39028616 PMCID: PMC11534315 DOI: 10.1093/neuonc/noae135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Glioblastoma is a highly aggressive brain cancer that is resistant to conventional immunotherapy strategies. Botensilimab, an Fc-enhanced anti-CTLA-4 antibody (FcE-aCTLA-4), has shown durable activity in "cold" and immunotherapy-refractory cancers. METHODS We evaluated the efficacy and immune microenvironment phenotype of a mouse analogue of FcE-aCTLA-4 in treatment-refractory preclinical models of glioblastoma, both as a monotherapy and in combination with doxorubicin delivered via low-intensity pulsed ultrasound and microbubbles (LIPU/MB). Additionally, we studied 4 glioblastoma patients treated with doxorubicin, anti-PD-1 with concomitant LIPU/MB to investigate the novel effect of doxorubicin modulating FcγR expressions in tumor-associated macrophages/microglia (TAMs). RESULTS FcE-aCTLA-4 demonstrated high-affinity binding to FcγRIV, the mouse ortholog of human FcγRIIIA, which was highly expressed in TAMs in human glioblastoma, most robustly at diagnosis. Notably, FcE-aCTLA-4-mediated selective depletion of intratumoral regulatory T cells (Tregs) via TAM-mediated phagocytosis, while sparing peripheral Tregs. Doxorubicin, a chemotherapeutic drug with immunomodulatory functions, was found to upregulate FcγRIIIA on TAMs in glioblastoma patients who received doxorubicin and anti-PD-1 with concomitant LIPU/MB. In murine models of immunotherapy-resistant gliomas, a combinatorial regimen of FcE-aCTLA-4, anti-PD-1, and doxorubicin with LIPU/MB, achieved a 90% cure rate, that was associated robust infiltration of activated CD8+ T cells and establishment of immunological memory as evidenced by rejection upon tumor rechallenge. CONCLUSIONS Our findings demonstrate that FcE-aCTLA-4 promotes robust immunomodulatory and anti-tumor effects in murine gliomas and is significantly enhanced when combined with anti-PD-1, doxorubicin, and LIPU/MB. We are currently investigating this combinatory strategy in a clinical trial (clinicaltrials.gov NCT05864534).
Collapse
Affiliation(s)
- Kwang-Soo Kim
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Karl Habashy
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andrew Gould
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Junfei Zhao
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, New York, USA
- Department of Systems Biology, Columbia University, New York, New York, USA
| | - Hinda Najem
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christina Amidei
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ruth Saganty
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Víctor A Arrieta
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Crismita Dmello
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Li Chen
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniel Y Zhang
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Brandyn Castro
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leah Billingham
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | | - Miguel Muzzio
- Life Science Group, IIT Research Institute (IITRI), Chicago, Illinois, USA
| | | | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Peng Zhang
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jason Miska
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Surya Padney
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Bin Zhang
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Raul Rabadan
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, New York, USA
- Department of Systems Biology, Columbia University, New York, New York, USA
| | - Joanna J Phillips
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Amy B Heimberger
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jian Hu
- Division of Basic Science Research, Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Roger Stupp
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Dhan Chand
- Agenus Inc., Lexington, Massachusetts, USA
| | - Catalina Lee-Chang
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adam M Sonabend
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
17
|
Zhu H, Allwin C, Bassous MG, Pouliopoulos AN. Focused ultrasound-mediated enhancement of blood-brain barrier permeability for brain tumor treatment: a systematic review of clinical trials. J Neurooncol 2024; 170:235-252. [PMID: 39207625 PMCID: PMC11538134 DOI: 10.1007/s11060-024-04795-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: 07/01/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE Brain tumors, particularly glioblastoma multiforme (GBM), present significant prognostic challenges despite multimodal therapies, including surgical resection, chemotherapy, and radiotherapy. One major obstacle is the limited drug delivery across the blood-brain barrier (BBB). Focused ultrasound (FUS) combined with systemically administered microbubbles has emerged as a non-invasive, targeted, and reversible approach to transiently open the BBB, thus enhancing drug delivery. This review examines clinical trials employing BBB opening techniques to optimise pharmacotherapy for brain tumors, evaluates current challenges, and proposes directions for further research. METHODS A systematic literature search was conducted in PubMed and ClinicalTrials.gov up to November 2023, searching for "ultrasound" AND "brain tumor". The search yielded 1446 results. After screening by title and abstract, followed by full-text screening (n = 48), 35 studies were included in the analysis. RESULTS Our analysis includes data from 11 published studies and 24 ongoing trials. The predominant focus of these studies is on glioma, including GMB and astrocytoma. One paper investigated brain metastasis from breast cancer. Evidence indicates that FUS facilitates BBB opening and enhances drug uptake following sonication. Exploration of FUS in the pediatric population is limited, with no published studies and only three ongoing trials dedicated to this demographic. CONCLUSION FUS is a promising strategy to safely disrupt the BBB, enabling precise and non-invasive lesion targeting, and enhance drug delivery. However, pharmacokinetic studies are required to quantitatively assess improvements in drug uptake. Most studies are phase I clinical trials, and long-term follow-up investigating patient outcomes is essential to evaluate the clinical benefit of this treatment approach. Further studies involving diverse populations and pathologies will be beneficial.
Collapse
Affiliation(s)
- Honglin Zhu
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Caitlin Allwin
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Monica G Bassous
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | | |
Collapse
|
18
|
Chen H, Koul D, Zhang Y, Ghobadi SN, Zhu Y, Hou Q, Chang E, Habte FG, Paulmurugan R, Khan S, Zheng Y, Graeber MB, Herschmann I, Lee KS, Wintermark M. Pulsed focused ultrasound alters the proteomic profile of the tumor microenvironment in a syngeneic mouse model of glioblastoma. J Neurooncol 2024; 170:347-361. [PMID: 39180641 DOI: 10.1007/s11060-024-04801-4] [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: 06/17/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE Glioblastoma (GBM), a lethal primary adult malignancy, is difficult to treat because of the restrictive nature of the blood-brain barrier (BBB), blood-tumor barrier (BTB), and the immunosuppressive tumor microenvironment (TME). Since pulsed focused ultrasound (pFUS) is currently used to improve therapeutic deliveries across these barriers, this study aims to characterize the impact of pFUS on the TME proteomics upon opening the BBB and BTB. METHODS We utilized MRI-guided, pFUS with ultrasound contrast microbubbles (termed 'pFUS' herein) to selectively and transiently open the BBB and BTB investigating proteomic modifications in the TME. Utilizing an orthotopically-allografted mouse GL26 GBM model (Ccr2RFP/wt - Cx3cr1GFP/wt), pFUS's effect on glioma proteomics was evaluated using a Luminex 48-plex assay. RESULTS pFUS treated tumors exhibited increases in pro-inflammatory cytokines, chemokines, and trophic factors (CCTFs). Proteomic changes in tumors tend to peak at 24 h after single pFUS session (1x), with levels then plateauing or declining over the subsequent 24 h. Tumors receiving three pFUS sessions (3x) showed elevated CCTFs levels peaking as early as 6 h after the third session. CONCLUSIONS pFUS together with microbubbles induces a sterile inflammatory response in the TME of a mouse GBM tumor. Moreover, this proinflammatory shift can be sustained and perhaps primed for more rapid responses upon multiple sessions of pFUS. These findings raise the intriguing potential that pFUS-induced BBB and BTB opening may not only be effective in facilitating the therapeutic agent delivery, but also be harnessed to modify the TME to assist immunotherapies in overcoming immune evasion in GBM.
Collapse
Affiliation(s)
- Hui Chen
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1482, Houston, TX, 77030, USA
| | - Dimpy Koul
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1482, Houston, TX, 77030, USA
| | - Yanrong Zhang
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA
| | - Sara Natasha Ghobadi
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA
| | - Yayu Zhu
- Salpointe Catholic High School, Tucson, AZ, USA
| | - Qingyi Hou
- Department of Radiology, Neuroradiology Division, Stanford University, Stanford, CA, USA
| | - Edwin Chang
- Department of Radiology, Molecular Imaging Program, Stanford University, Stanford, CA, USA
| | - Frezghi G Habte
- Department of Radiology, Molecular Imaging Program, Stanford University, Stanford, CA, USA
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program, Stanford University, Stanford, CA, USA
| | - Sabbir Khan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuqi Zheng
- Ken Parker Brain Tumour Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Manuel B Graeber
- Ken Parker Brain Tumour Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2050, Australia
- University of Sydney Association of Professors (USAP), University of Sydney, Camperdown, NSW, 2006, Australia
| | - Iris Herschmann
- The Human Immune Monitoring Center (HIMC), Stanford University, Stanford, CA, USA
| | - Kevin S Lee
- Departments of Neuroscience and Neurosurgery, Center for Brain Immunology and Glia, School of Medicine, University of Virginia, 409 Lane Road, MR4 Building, PO Box 801392, Charlottesville, VA, 22903, USA.
| | - Max Wintermark
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1482, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Mauser A, Waibel I, Banerjee K, Mujeeb AA, Gan J, Lee S, Brown W, Lang N, Gregory J, Raymond J, Franzeb M, Schwendeman A, Castro MG, Lahann J. Controlled Delivery of Paclitaxel via Stable Synthetic Protein Nanoparticles. ADVANCED THERAPEUTICS 2024; 7:2400208. [PMID: 39575154 PMCID: PMC11580025 DOI: 10.1002/adtp.202400208] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Indexed: 11/24/2024]
Abstract
Despite decades of intense research, glioma remains a disease for which no adequate clinical treatment exists. Given the ongoing therapeutic failures of conventional treatment approaches, nanomedicine may offer alternative options because it can increase the bioavailability of drugs and alter their pharmacokinetics. Here, a new type of synthetic protein nanoparticles (SPNPs) is reported that allow for effective loading and controlled release of the potent cancer drug, paclitaxel (PTX) - a drug that so far has been unsuccessful in glioma treatment due to hydrophobicity, low solubility, and associated delivery challenges. SPNPs are prepared by electrohydrodynamic (EHD) jetting of dilute solutions of PTX-loaded albumin made by high-pressure homogenization. After EHD jetting, PTX SPNPs possess a dry diameter of 165 ± 44 nm, hydrated diameter of 297 ± 102 nm, and a zeta potential of -19 ± 8 mV in water. For the SPNP formulation with a total PTX loading of 9.4%, the loading efficiency is 94%, and controlled release of PTX is observed over two weeks (6% burst release). PTX SPNPs are more potent (68% lethality) than free PTX (45% lethality using 0.2% dimethyl sulfoxide). PTX SPNPs in combination with IR show a significant survival benefit in glioma-bearing mouse models, avoid adverse liver toxicity, and maintain a normal brain architecture. Immunohistochemistry reveals a dramatic tumor size reduction including 40% long-term survivors without discernible signs of tumor. Using flexibly engineered SPNPs, this work outlines an efficient strategy for the delivery of hydrophobic drugs that are otherwise notoriously hard to deliver.
Collapse
Affiliation(s)
- Ava Mauser
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Isabel Waibel
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical and Process Engineering, Karlsruhe Institute of Technology, 76344 Karlsruhe, Germany
| | - Kaushik Banerjee
- Department of Neurosurgery and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anzar A Mujeeb
- Department of Neurosurgery and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jingyao Gan
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sophia Lee
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - William Brown
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nigel Lang
- Department of Neurosurgery and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jason Gregory
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeffery Raymond
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthias Franzeb
- Department of Chemical and Process Engineering, Karlsruhe Institute of Technology, 76344 Karlsruhe, Germany
| | - Anna Schwendeman
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria G Castro
- Department of Neurosurgery and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joerg Lahann
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
20
|
Farzad Maroufi S, Sadegh Fallahi M, Parmis Maroufi S, Sheehan JP. Focused ultrasound blood-brain barrier disruption in high-grade gliomas: Scoping review of clinical studies. J Clin Neurosci 2024; 128:110786. [PMID: 39146579 DOI: 10.1016/j.jocn.2024.110786] [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: 05/24/2024] [Revised: 08/01/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND This scoping review aims to comprehensively review the available literature on the safety and efficacy of focused ultrasound (FUS) for blood-brain barrier disruption (BBBD) in patients with high-grade gliomas, including glioblastoma (GBM). High-grade gliomas pose significant challenges in neuro-oncology due to their aggressiveness and intricate location, often limiting the efficacy of traditional treatments. FUS offers a promising approach by transiently disrupting the blood-brain barrier, thereby facilitating enhanced drug delivery to tumor cells while minimizing systemic side effects. METHODS A scoping review adhering to PRISMA guidelines was conducted to explore the literature on FUS-induced BBBD in glioma patients. PubMed and Embase databases were searched from inception to April 2024 using defined keywords. Original clinical studies focusing on FUS for BBBD in gliomas were included. Two reviewers independently screened records, with conflicts resolved by a third reviewer. Data extraction and quality assessment were performed accordingly. RESULTS A total of 1,310 studies were initially identified, resulting in nine eligible studies after screening and selection. These studies, published between 2016 and 2024, included 106 patients (39.6 % female) with ages ranging from 29 to 80 years. Recurrent GBM was the most common diagnosis (100 patients), with other diagnoses including anaplastic astrocytoma, diffuse infiltrating glioma, and oligodendroglioma. Various FUS devices and microbubble contrast agents were employed across the studies. Safety and efficacy were assessed in both experimental and clinical settings, with no significant adverse events reported during BBBD procedures. Notably, BBBD facilitated enhanced drug delivery to tumor tissue, demonstrating potential therapeutic benefits. CONCLUSION Studies investigating BBBD using FUS demonstrate promising outcomes in experimental and clinical settings. BBBD procedures in patients with malignant gliomas and recurrent GBM show safety and successful enhancement of drug delivery potential. Overall, FUS-mediated BBBD emerges as a safe and feasible approach for improving therapeutic outcomes in brain tumor patients, warranting further clinical exploration and optimization.
Collapse
Affiliation(s)
- Seyed Farzad Maroufi
- Neurosurgical Research Network (NRN), Universal Scientific Education and Research Network (USERN), Tehran University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sadegh Fallahi
- Neurosurgical Research Network (NRN), Universal Scientific Education and Research Network (USERN), Tehran University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyede Parmis Maroufi
- Neurosurgical Research Network (NRN), Universal Scientific Education and Research Network (USERN), Tehran University of Medical Sciences, Tehran, Iran
| | - Jason P Sheehan
- Department of Neurological Surgery, University of Virginia, Charlottesville, USA.
| |
Collapse
|
21
|
Piper K, Kumar JI, Domino J, Tuchek C, Vogelbaum MA. Consensus review on strategies to improve delivery across the blood-brain barrier including focused ultrasound. Neuro Oncol 2024; 26:1545-1556. [PMID: 38770775 PMCID: PMC11376463 DOI: 10.1093/neuonc/noae087] [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: 05/21/2024] [Indexed: 05/22/2024] Open
Abstract
Drug delivery to the central nervous system (CNS) has been a major challenge for CNS tumors due to the impermeability of the blood-brain barrier (BBB). There has been a multitude of techniques aimed at overcoming the BBB obstacle aimed at utilizing natural transport mechanisms or bypassing the BBB which we review here. Another approach that has generated recent interest in the recently published literature is to use new technologies (Laser Interstitial Thermal Therapy, LITT; or Low-Intensity Focused Ultrasound, LIFU) to temporarily increase BBB permeability. This review overviews the advantages, disadvantages, and major advances of each method. LIFU has been a major area of research to allow for chemotherapeutics to cross the BBB which has a particular emphasis in this review. While most of the advances remain in animal studies, there are an increasing number of translational clinical trials that will have results in the next few years.
Collapse
Affiliation(s)
- Keaton Piper
- Department of Neurosurgery, University of South Florida, Tampa, Florida, USA
| | - Jay I Kumar
- Department of Neurosurgery, University of South Florida, Tampa, Florida, USA
| | - Joseph Domino
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Chad Tuchek
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Michael A Vogelbaum
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| |
Collapse
|
22
|
Madani F, Morovvati H, Webster TJ, Najaf Asaadi S, Rezayat SM, Hadjighassem M, Khosravani M, Adabi M. Combination chemotherapy via poloxamer 188 surface-modified PLGA nanoparticles that traverse the blood-brain-barrier in a glioblastoma model. Sci Rep 2024; 14:19516. [PMID: 39174603 PMCID: PMC11341868 DOI: 10.1038/s41598-024-69888-1] [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: 03/08/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024] Open
Abstract
The effect of chemotherapy for anti-glioblastoma is limited due to insufficient drug delivery across the blood-brain-barrier. Poloxamer 188-coated nanoparticles can enhance the delivery of nanoparticles across the blood-brain-barrier. This study presents the design, preparation, and evaluation of a combination of PLGA nanoparticles (PLGA NPs) loaded with methotrexate (P-MTX NPs) and PLGA nanoparticles loaded with paclitaxel (P-PTX NPs), both of which were surface-modified with poloxamer188. Cranial tumors were induced by implanting C6 cells in a rat model and MRI demonstrated that the tumors were indistinguishable in the two rats with P-MTX NPs + P-PTX NPs treated groups. Brain PET scans exhibited a decreased brain-to-background ratio which could be attributed to the diminished metabolic tumor volume. The expression of Ki-67 as a poor prognosis factor, was significantly lower in P-MTX NPs + P-PTX NPs compared to the control. Furthermore, the biodistribution of PLGA NPs was determined by carbon quantum dots loaded into PLGA NPs (P-CQD NPs), and quantitative analysis of ex-vivo imaging of the dissected organs demonstrated that 17.2 ± 0.6% of the NPs were concentrated in the brain after 48 h. The findings highlight the efficacy of combination nanochemotherapy in glioblastoma treatment, indicating the need for further preclinical studies.
Collapse
Affiliation(s)
- Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Morovvati
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Thomas J Webster
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, China
- Program in Materials Science, UFPI, Teresina, Brazil
| | - Sareh Najaf Asaadi
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Seyed Mahdi Rezayat
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Miao K, Xia X, Zou Y, Shi B. Small Scale, Big Impact: Nanotechnology-Enhanced Drug Delivery for Brain Diseases. Mol Pharm 2024; 21:3777-3799. [PMID: 39038108 DOI: 10.1021/acs.molpharmaceut.4c00387] [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: 07/24/2024]
Abstract
Central nervous system (CNS) diseases, ranging from brain cancers to neurodegenerative disorders like dementia and acute conditions such as strokes, have been heavily burdening healthcare and have a direct impact on patient quality of life. A significant hurdle in developing effective treatments is the presence of the blood-brain barrier (BBB), a highly selective barrier that prevents most drugs from reaching the brain. The tight junctions and adherens junctions between the endothelial cells and various receptors expressed on the cells make the BBB form a nonfenestrated and highly selective structure that is crucial for brain homeostasis but complicates drug delivery. Nanotechnology offers a novel pathway to circumvent this barrier, with nanoparticles engineered to ferry drugs across the BBB, protect drugs from degradation, and deliver medications to the designated area. After years of development, nanoparticle optimization, including sizes, shapes, surface modifications, and targeting ligands, can enable nanomaterials tailored to specific brain drug delivery settings. Moreover, smart nano drug delivery systems can respond to endogenous and exogenous stimuli that control subsequent drug release. Here, we address the importance of the BBB in brain disease treatment, summarize different delivery routes for brain drug delivery, discuss the cutting-edge nanotechnology-based strategies for brain drug delivery, and further offer valuable insights into how these innovations in nanoparticle technology could revolutionize the treatment of CNS diseases, presenting a promising avenue for noninvasive, targeted therapeutic interventions.
Collapse
Affiliation(s)
- Kaiting Miao
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Xue Xia
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Yan Zou
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Bingyang Shi
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
24
|
Montero AS, Aliouat I, Ribon M, Canney M, Goldwirt L, Mourah S, Berriat F, Lobsiger CS, Pradat PF, Salachas F, Bruneteau G, Carpentier A, Boillée S. Effect of ultrasound-mediated blood-spinal cord barrier opening on survival and motor function in females in an amyotrophic lateral sclerosis mouse model. EBioMedicine 2024; 106:105235. [PMID: 38996764 PMCID: PMC11284947 DOI: 10.1016/j.ebiom.2024.105235] [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: 01/31/2023] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by a progressive loss of motor neurons. The limited efficacy of recent therapies in clinical development may be linked to lack of drug penetration to the affected motor neurons due to the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB). METHODS In this work, the safety and efficacy of repeated short transient opening of the BSCB by low intensity pulsed ultrasound (US, sonication) was studied in females of an ALS mouse model (B6.Cg-Tg(SOD1∗G93A)1Gur/J). The BSCB was disrupted using a 1 MHz ultrasound transducer coupled to the spinal cord, with and without injection of insulin-like growth factor 1 (IGF1), a neurotrophic factor that has previously shown efficacy in ALS models. FINDINGS Results in wild-type (WT) animals demonstrated that the BSCB can be safely disrupted and IGF1 concentrations significantly enhanced after a single session of transient BSCB disruption (176 ± 32 μg/g vs. 0.16 ± 0.008 μg/g, p < 0.0001). Five repeated weekly US sessions performed in female ALS mice demonstrated a survival advantage in mice treated with IGF1 and US (US IGF1) compared to treatment with IGF1 alone (176 vs. 166 days, p = 0.0038). Surprisingly, this survival advantage was also present in mice treated with US alone vs. untreated mice (178.5 vs. 166.5 days, p = 0.0061). Muscle strength did not show difference among the groups. Analysis of glial cell immunoreactivity and microglial transcriptome showing reduced cell proliferation pathways, in addition to lymphocyte infiltration, suggested that the beneficial effect of US or US IGF1 could act through immune cell modulation. INTERPRETATION These results show the first step towards a possible beneficial impact of transient BSCB opening for ALS therapy and suggest implication of immune cells. FUNDING Fondation pour la Recherche Médicale (FRM). Investissements d'avenirANR-10-IAIHU-06, Société Française de Neurochirurgie (SFNC), Fond d'étude et de Recherche du Corps Medical (FERCM), Aide à la Recherche des Maladies du Cerveau (ARMC), SLA Fondation Recherche (SLAFR), French Ministry for High Education and Research (MENR), Carthera, Laboratoire de Recherche en Technologies Chirurgicales Avancées (LRTCA).
Collapse
Affiliation(s)
- Anne-Sophie Montero
- Sorbonne Université, Neurosurgery Department, AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Advanced Surgical Research Technology Laboratory, Paris, France; Sorbonne Université, GRC 23, Brain Machine Interface, AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Ilyes Aliouat
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Matthieu Ribon
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | | | - Lauriane Goldwirt
- AP-HP, Pharmacology Department, Hôpital de Saint-Louis, Paris, France
| | - Samia Mourah
- AP-HP, Pharmacology Department, Hôpital de Saint-Louis, Paris, France
| | - Félix Berriat
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Christian S Lobsiger
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Pierre-François Pradat
- AP-HP, Centre de Reference Maladie Rare SLA, Neurology Department, Pitié-Salpêtrière Hospital, Paris, France
| | - François Salachas
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France; AP-HP, Centre de Reference Maladie Rare SLA, Neurology Department, Pitié-Salpêtrière Hospital, Paris, France
| | - Gaëlle Bruneteau
- AP-HP, Centre de Reference Maladie Rare SLA, Neurology Department, Pitié-Salpêtrière Hospital, Paris, France
| | - Alexandre Carpentier
- Sorbonne Université, Neurosurgery Department, AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Advanced Surgical Research Technology Laboratory, Paris, France; Sorbonne Université, GRC 23, Brain Machine Interface, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Séverine Boillée
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
25
|
Zou Y, Xu L, Wang W, Zhu X, Lin J, Li H, Chen J, Xu W, Gao H, Wu X, Yin Z, Wang Q. Muscone restores anoikis sensitivity in TMZ-resistant glioblastoma cells by suppressing TOP2A via the EGFR/Integrin β1/FAK signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155714. [PMID: 38723526 DOI: 10.1016/j.phymed.2024.155714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/15/2024] [Accepted: 05/04/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Temozolomide (TMZ) resistance is the main obstacle faced by glioblastoma multiforme (GBM) treatment. Muscone, one of the primary active pharmacological ingredients of Shexiang (Moschus), can cross the blood-brain barrier (BBB) and is being investigated as an antineoplastic medication. However, muscone treatment for GBM has received little research, and its possible mechanisms are still unclear. PURPOSE This study aims to evaluate the effect and the potential molecular mechanism of muscone on TMZ-resistant GBM cells. METHODS The differentially expressed genes (DEGs) between TMZ-resistant GBM cells and TMZ-sensitive GBM cells were screened using GEO2R. By progressively raising the TMZ concentration, a relatively stable TMZ-resistant human GBM cell line was established. The drug-resistance traits of U251-TR cells were assessed via the CCK-8 assay and Western Blot analysis of MGMT and TOP2A expression. Cell viability, cell proliferation, cell migration ability, and drug synergism were detected by the CCK-8 assay, colony formation assay, wound healing assay, and drug interaction relationship test, respectively. Anoikis was quantified by Calcein-AM/EthD-1 staining, MTT assay, and flow cytometry. Measurements of cell cycle arrest, apoptosis, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) were performed using cell cycle staining, Annexin V-FITC/PI labeling, JC-1 assay, and ROS assay, respectively. DNA damage was measured by TUNEL assay, alkaline comet assay, and γ-H2AX foci assay. GEPIA was used to investigate the link between the anoikis marker (FAK)/drug resistance gene and critical proteins in the EGFR/Integrin β1 signaling pathway. Molecular docking was used to anticipate the probable targets of muscone. The intracellular co-localization and expression of EGFR and FAK were shown using immunofluorescence. The U251-TR cell line stably overexpressing EGFR was constructed using lentiviral transduction to assess the involvement of EGFR-related signaling in anoikis resistance. Western Blot was employed to detect the expression of migration-related proteins, cyclins, anoikis-related proteins, DNA damage/repair-related proteins, and associated pathway proteins. RESULTS DEGs analysis identified 97 deregulated chemotherapy-resistant genes and 3779 upregulated genes in TMZ-resistant GBM cells. Subsequent experiments verified TMZ resistance and the hyper-expression of DNA repair-related genes (TOP2A and MGMT) in continuously low-dose TMZ-induced U251-TR cells. Muscone exhibited dose-dependent inhibition of U251-TR cell migration and proliferation, and its co-administration with TMZ showed the potential for enhanced therapeutic efficacy. By downregulating FAK, muscone reduced anoikis resistance in anchorage-independent U251-TR cells. It also caused cell cycle arrest in the G2/M phase by upregulating p21 and downregulating CDK1, CDK2, and Cyclin E1. Muscone-induced anoikis was accompanied by mitochondrial membrane potential collapse, ROS production, an increase in the BAX/Bcl-2 ratio, as well as elevated levels of Cytochrome c (Cyt c), cleaved caspase-9, and cleaved caspase-3. These findings indicated that muscone might trigger mitochondrial-dependent anoikis via ROS generation. Moreover, significant DNA damage, DNA double-strand breaks (DSBs), the formation of γ-H2AX foci, and a reduction in TOP2A expression are also associated with muscone-induced anoikis. Overexpression of EGFR in U251-TR cells boosted the expression of Integrin β1, FAK, β-Catenin, and TOP2A, whereas muscone suppressed the expression levels of EGFR, Integrin β1, β-Catenin, FAK, and TOP2A. Muscone may influence the expression of the key DNA repair enzyme, TOP2A, by suppressing the EGFR/Integrin β1/FAK pathway. CONCLUSION We first demonstrated that muscone suppressed TOP2A expression through the EGFR/Integrin β1/FAK pathway, hence restoring anoikis sensitivity in TMZ-resistant GBM cells. These data suggest that muscone may be a promising co-therapeutic agent for enhancing GBM treatment, particularly in cases of TMZ-resistant GBM with elevated TOP2A expression.
Collapse
Affiliation(s)
- Yuheng Zou
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lanyang Xu
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Wanyu Wang
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiao Zhu
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiaqi Lin
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Huazhao Li
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiali Chen
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Wei Xu
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Haiqiong Gao
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xianghui Wu
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhixin Yin
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qirui Wang
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
26
|
Sarkar S, Greer J, Marlowe NJ, Medvid A, Ivan ME, Kolishetti N, Dhar S. Stemness, invasion, and immunosuppression modulation in recurrent glioblastoma using nanotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1976. [PMID: 39091260 DOI: 10.1002/wnan.1976] [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: 08/07/2023] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 08/04/2024]
Abstract
The recurrent nature of glioblastoma negatively impacts conventional treatment strategies leading to a growing need for nanomedicine. Nanotherapeutics, an approach designed to deliver drugs to specific sites, is experiencing rapid growth and gaining immense popularity. Having potential in reaching the hard-to-reach disease sites, this field has the potential to show high efficacy in combatting glioblastoma progression. The presence of glioblastoma stem cells (GSCs) is a major factor behind the poor prognosis of glioblastoma multiforme (GBM). Stemness potential, heterogeneity, and self-renewal capacity, are some of the properties that make GSCs invade across the distant regions of the brain. Despite advances in medical technology and MRI-guided maximal surgical resection, not all GSCs residing in the brain can be removed, leading to recurrent disease. The aggressiveness of GBM is often correlated with immune suppression, where the T-cells are unable to infiltrate the cancer initiating GSCs. Standard of care therapies, including surgery and chemotherapy in combination with radiation therapy, have failed to tackle all the challenges of the GSCs, making it increasingly important for researchers to develop strategies to tackle their growth and proliferation and reduce the recurrence of GBM. Here, we will focus on the advancements in the field of nanomedicine that has the potential to show positive impact in managing glioblastoma tumor microenvironment. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Shrita Sarkar
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Jessica Greer
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Nathaniel J Marlowe
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Angeline Medvid
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Michael E Ivan
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Nagesh Kolishetti
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, Florida, USA
- Herbert Wertheim College of Medicine, Institute of Neuroimmune Pharmacology, Miami, Florida International University, Florida, USA
| | - Shanta Dhar
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Chemistry, University of Miami, Coral Gables, Florida, USA
| |
Collapse
|
27
|
Ma C, Ye Q, Qian K, Dai M, Gan L, Yang J, Jia Z, Pan Z, Cai Q, Jiang T, Lin X. Anti-glioma effect of paclitaxel mediated by specific mode electroacupuncture stimulation and the related role of the Hedgehog pathway. Brain Res Bull 2024; 213:110985. [PMID: 38806118 DOI: 10.1016/j.brainresbull.2024.110985] [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: 04/04/2024] [Revised: 05/11/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
INTRODUCTION Paclitaxel (PTX) cannot effectively treat glioma because it cannot cross the bloodbrain barrier (BBB). A specific mode electroacupuncture stimulation (SMES) can temporarily open the BBB, thereby improving drug delivery to the brain. This study aimed to observe SMES-mediated accumulation of PTX in the brain and its anti-glioma effect and explore the role of the Hedgehog pathway. METHODS The acupoint selectivity of SMES in opening the BBB was examined in normal rats. The penetration and anti-glioma activity were determined in a C6-Luc glioma rat model. SMES was performed using 2/100 Hz, 3 mA, 6-6 s, and 40 min The survival curve was analysed by the KaplanMeier method, brain tumour pathology and size was observed by HE staining, and in vivo imaging system respectively. RESULTS SMES-induced BBB opening had acupoint selectivity. SMES could improve PTX accumulation in brain and SMES-mediated PTX delivery showed enhanced anti-glioma activity due to better brain penetration. Hedgehog pathway was involved in SMES-mediated PTX delivery by regulating Occludin expression. CONCLUSION SMES at the head acupoints to deliver PTX is a feasible and effective method for treating glioma. The Hedgehog pathway may play a key role in SMES-mediated PTX delivery across the BBB.
Collapse
Affiliation(s)
- Congcong Ma
- Zhejiang Chinese Medical University Affiliated Third Hospital, China; The Third Clinical Medical College, Zhejiang Chinese Medical University, China; Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Qinyu Ye
- The Third Clinical Medical College, Zhejiang Chinese Medical University, China
| | - Kecheng Qian
- The Third Clinical Medical College, Zhejiang Chinese Medical University, China
| | - Mengyuan Dai
- The Third Clinical Medical College, Zhejiang Chinese Medical University, China
| | - Lin Gan
- The Third Clinical Medical College, Zhejiang Chinese Medical University, China
| | - Jinding Yang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, China
| | - Zhaoxing Jia
- The Third Clinical Medical College, Zhejiang Chinese Medical University, China
| | - Zixin Pan
- The Third Clinical Medical College, Zhejiang Chinese Medical University, China
| | - Qian Cai
- The Third Clinical Medical College, Zhejiang Chinese Medical University, China
| | - Tianxiang Jiang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, China
| | - Xianming Lin
- Zhejiang Chinese Medical University Affiliated Third Hospital, China; The Third Clinical Medical College, Zhejiang Chinese Medical University, China; Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
28
|
Arrieta VA, Gould A, Kim KS, Habashy KJ, Dmello C, Vázquez-Cervantes GI, Palacín-Aliana I, McManus G, Amidei C, Gomez C, Dhiantravan S, Chen L, Zhang DY, Saganty R, Cholak ME, Pandey S, McCord M, McCortney K, Castro B, Ward R, Muzzio M, Bouchoux G, Desseaux C, Canney M, Carpentier A, Zhang B, Miska JM, Lesniak MS, Horbinski CM, Lukas RV, Stupp R, Lee-Chang C, Sonabend AM. Ultrasound-mediated delivery of doxorubicin to the brain results in immune modulation and improved responses to PD-1 blockade in gliomas. Nat Commun 2024; 15:4698. [PMID: 38844770 PMCID: PMC11156895 DOI: 10.1038/s41467-024-48326-w] [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: 03/08/2023] [Accepted: 04/29/2024] [Indexed: 06/09/2024] Open
Abstract
Given the marginal penetration of most drugs across the blood-brain barrier, the efficacy of various agents remains limited for glioblastoma (GBM). Here we employ low-intensity pulsed ultrasound (LIPU) and intravenously administered microbubbles (MB) to open the blood-brain barrier and increase the concentration of liposomal doxorubicin and PD-1 blocking antibodies (aPD-1). We report results on a cohort of 4 GBM patients and preclinical models treated with this approach. LIPU/MB increases the concentration of doxorubicin by 2-fold and 3.9-fold in the human and murine brains two days after sonication, respectively. Similarly, LIPU/MB-mediated blood-brain barrier disruption leads to a 6-fold and a 2-fold increase in aPD-1 concentrations in murine brains and peritumoral brain regions from GBM patients treated with pembrolizumab, respectively. Doxorubicin and aPD-1 delivered with LIPU/MB upregulate major histocompatibility complex (MHC) class I and II in tumor cells. Increased brain concentrations of doxorubicin achieved by LIPU/MB elicit IFN-γ and MHC class I expression in microglia and macrophages. Doxorubicin and aPD-1 delivered with LIPU/MB results in the long-term survival of most glioma-bearing mice, which rely on myeloid cells and lymphocytes for their efficacy. Overall, this translational study supports the utility of LIPU/MB to potentiate the antitumoral activities of doxorubicin and aPD-1 for GBM.
Collapse
Affiliation(s)
- Víctor A Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Andrew Gould
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kwang-Soo Kim
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karl J Habashy
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Crismita Dmello
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gustavo I Vázquez-Cervantes
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Irina Palacín-Aliana
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Deparment of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Graysen McManus
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Christina Amidei
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Cristal Gomez
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Silpol Dhiantravan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Li Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Daniel Y Zhang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ruth Saganty
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Meghan E Cholak
- Department of Medicine, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Surya Pandey
- Department of Medicine, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Matthew McCord
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Deparment of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Brandyn Castro
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Rachel Ward
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Miguel Muzzio
- Life Sciences Group, IIT Research Institute, Chicago, IL, USA
| | | | | | | | - Alexandre Carpentier
- Sorbonne Université, Inserm, CNRS, UMR S 1127, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Service de Neurochirurgie, Paris, France
| | - Bin Zhang
- Department of Medicine, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jason M Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Craig M Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Rimas V Lukas
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Roger Stupp
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Medicine, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
29
|
Seas AA, Malla AP, Sharifai N, Winkles JA, Woodworth GF, Anastasiadis P. Microbubble-Enhanced Focused Ultrasound for Infiltrating Gliomas. Biomedicines 2024; 12:1230. [PMID: 38927437 PMCID: PMC11200892 DOI: 10.3390/biomedicines12061230] [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: 05/03/2024] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Infiltrating gliomas are challenging to treat, as the blood-brain barrier significantly impedes the success of therapeutic interventions. While some clinical trials for high-grade gliomas have shown promise, patient outcomes remain poor. Microbubble-enhanced focused ultrasound (MB-FUS) is a rapidly evolving technology with demonstrated safety and efficacy in opening the blood-brain barrier across various disease models, including infiltrating gliomas. Initially recognized for its role in augmenting drug delivery, the potential of MB-FUS to augment liquid biopsy and immunotherapy is gaining research momentum. In this review, we will highlight recent advancements in preclinical and clinical studies that utilize focused ultrasound to treat gliomas and discuss the potential future uses of image-guided precision therapy using focused ultrasound.
Collapse
Affiliation(s)
- Alexandra A. Seas
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Adarsha P. Malla
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nima Sharifai
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD 21201, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeffrey A. Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD 21201, USA
| | - Graeme F. Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD 21201, USA
| | - Pavlos Anastasiadis
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD 21201, USA
| |
Collapse
|
30
|
Li S, Meng C, Hao Q, Zhou R, Dai L, Guo Y, Zhao S, Zhou X, Lou C, Xu J, Xu P, Yang J, Ding Y, Lv Y, Han S, Li S, Li J, Kang H, Xiao Z, Tan M, Ma X, Wu H. "On/off"-switchable crosslinked PTX-nanoformulation with improved precise delivery for NSCLC brain metastases and restrained adverse reaction over nab-PTX. Biomaterials 2024; 307:122537. [PMID: 38492523 DOI: 10.1016/j.biomaterials.2024.122537] [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: 01/13/2024] [Revised: 02/29/2024] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
Non-small cell lung cancer (NSCLC) brain metastases present a significant treatment challenge due to limited drug delivery efficiency and severe adverse reactions. In this study, we address these challenges by designing a "on/off" switchable crosslinked paclitaxel (PTX) nanocarrier, BPM-PD, with novel ultra-pH-sensitive linkages (pH 6.8 to 6.5). BPM-PD demonstrates a distinct "on/off" switchable release of the anti-cancer drug paclitaxel (PTX) in response to the acidic extratumoral microenvironment. The "off" state of BPM-PD@PTX effectively prevents premature drug release in the blood circulation, blood-brain barrier (BBB)/blood-tumor barrier (BTB), and normal brain tissue, surpassing the clinical PTX-nanoformulation (nab-PTX). Meanwhile, the "on" state facilitates precise delivery to NSCLC brain metastases cells. Compared to nab-PTX, BPM-PD@PTX demonstrates improved therapeutic efficacy with a reduced tumor area (only 14.6%) and extended survival duration, while mitigating adverse reactions (over 83.7%) in aspartate aminotransferase (AST) and alanine aminotransferase (ALT), offering a promising approach for the treatment of NSCLC brain metastases. The precise molecular switch also helped to increase the PTX maximum tolerated dose from 25 mg/kg to 45 mg/kg This research contributes to the field of cancer therapeutics and has significant implications for improving the clinical outcomes of NSCLC patients.
Collapse
Affiliation(s)
- Shuaijun Li
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China; School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Caiting Meng
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qian Hao
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ruina Zhou
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Luyao Dai
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yucheng Guo
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Sitong Zhao
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xin Zhou
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Chunju Lou
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ji Xu
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Peng Xu
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jinfan Yang
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an, Shaanxi, 710021, China
| | - Yifan Ding
- School of Pharmacy, Health Science Center of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yanni Lv
- School of Pharmacy, Health Science Center of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Shengli Han
- School of Pharmacy, Health Science Center of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Shuai Li
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jing Li
- Xijing Hypertrophic Cardiomyopathy Center, Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, Xian, Shaanxi, China
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zhengtao Xiao
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, Liaoning, 116034, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China; School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Hao Wu
- Department of Oncology, The Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China; School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
31
|
Conq J, Joudiou N, Préat V, Gallez B. Exploring the Impact of Irradiation on Glioblastoma Blood-Brain-Barrier Permeability: Insights from Dynamic-Contrast-Enhanced-MRI and Histological Analysis. Biomedicines 2024; 12:1091. [PMID: 38791053 PMCID: PMC11118616 DOI: 10.3390/biomedicines12051091] [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: 04/08/2024] [Revised: 04/26/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: Glioblastoma (GB) presents a formidable challenge in neuro-oncology due to its aggressive nature, limited treatment options, and poor prognosis. The blood-brain barrier (BBB) complicates treatment by hindering drug delivery to the tumor site, particularly to the infiltrative cells in the margin of the tumor, which are mainly responsible for tumor recurrence. Innovative strategies are therefore needed to enhance drug delivery in the margins of the tumor. This study explores whether irradiation can enhance BBB permeability by assessing hemodynamic changes and the distribution of contrast agents in the core and the margins of GB tumors. (2) Methods: Mice grafted with U-87MG cells were exposed to increasing irradiation doses. The distribution of contrast agents and hemodynamic parameters was evaluated using both non-invasive magnetic resonance imaging (MRI) techniques with gadolinium-DOTA as a contrast agent and invasive histological analysis with Evans blue, a fluorescent vascular leakage marker. Diffusion-MRI was also used to assess cytotoxic effects. (3) Results: The histological study revealed a complex dose-dependent effect of irradiation on BBB integrity, with increased vascular leakage at 5 Gy but reduced leakage at higher doses (10 and 15 Gy). However, there was no significant increase in the diffusion of Gd-DOTA outside the tumor area by MRI. (4) Conclusions: The increase in BBB permeability could be an interesting approach to enhance drug delivery in glioblastoma margins for low irradiation doses. In this model, DCE-MRI analysis was of limited value in assessing the BBB opening in glioblastoma after irradiation.
Collapse
Affiliation(s)
- Jérôme Conq
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Advanced Drug Delivery and Biomaterials Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Nicolas Joudiou
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| |
Collapse
|
32
|
Mao M, Wu Y, He Q. Recent advances in targeted drug delivery for the treatment of glioblastoma. NANOSCALE 2024; 16:8689-8707. [PMID: 38606460 DOI: 10.1039/d4nr01056f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Glioblastoma multiforme (GBM) is one of the highly malignant brain tumors characterized by significant morbidity and mortality. Despite the recent advancements in the treatment of GBM, major challenges persist in achieving controlled drug delivery to tumors. The management of GBM poses considerable difficulties primarily due to unresolved issues in the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB) and GBM microenvironment. These factors limit the uptake of anti-cancer drugs by the tumor, thus limiting the therapeutic options. Current breakthroughs in nanotechnology provide new prospects concerning unconventional drug delivery approaches for GBM treatment. Specifically, swimming nanorobots show great potential in active targeted delivery, owing to their autonomous propulsion and improved navigation capacities across biological barriers, which further facilitate the development of GBM-targeted strategies. This review presents an overview of technological progress in different drug administration methods for GBM. Additionally, the limitations in clinical translation and future research prospects in this field are also discussed. This review aims to provide a comprehensive guideline for researchers and offer perspectives on further development of new drug delivery therapies to combat GBM.
Collapse
Affiliation(s)
- Meng Mao
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Yingjie Wu
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Qiang He
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
33
|
Perolina E, Meissner S, Raos B, Harland B, Thakur S, Svirskis D. Translating ultrasound-mediated drug delivery technologies for CNS applications. Adv Drug Deliv Rev 2024; 208:115274. [PMID: 38452815 DOI: 10.1016/j.addr.2024.115274] [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: 09/28/2023] [Revised: 02/18/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024]
Abstract
Ultrasound enhances drug delivery into the central nervous system (CNS) by opening barriers between the blood and CNS and by triggering release of drugs from carriers. A key challenge in translating setups from in vitro to in vivo settings is achieving equivalent acoustic energy delivery. Multiple devices have now been demonstrated to focus ultrasound to the brain, with concepts emerging to also target the spinal cord. Clinical trials to date have used ultrasound to facilitate the opening of the blood-brain barrier. While most have focused on feasibility and safety considerations, therapeutic benefits are beginning to emerge. To advance translation of these technologies for CNS applications, researchers should standardise exposure protocol and fine-tune ultrasound parameters. Computational modelling should be increasingly used as a core component to develop both in vitro and in vivo setups for delivering accurate and reproducible ultrasound to the CNS. This field holds promise for transformative advancements in the management and pharmacological treatment of complex and challenging CNS disorders.
Collapse
Affiliation(s)
- Ederlyn Perolina
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Svenja Meissner
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Brad Raos
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Bruce Harland
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Sachin Thakur
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand.
| |
Collapse
|
34
|
Bakker A, Ixkes AE, Venugopal H, Ries MG, Lak NSM, de Vos FYFL, van Vuurden DG, Snijders TJ. Focused Ultrasound-Enhanced Liquid Biopsy: A Promising Diagnostic Tool for Brain Tumor Patients. Cancers (Basel) 2024; 16:1576. [PMID: 38672658 PMCID: PMC11049441 DOI: 10.3390/cancers16081576] [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: 03/18/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
The performance of minimally invasive molecular diagnostic tools in brain tumors, such as liquid biopsy, has so far been limited by the blood-brain barrier (BBB). The BBB hinders the release of brain tumor biomarkers into the bloodstream. The use of focused ultrasound in conjunction with microbubbles has been shown to temporarily open the BBB (FUS-BBBO). This may enhance blood-based tumor biomarker levels. This systematic review provides an overview of the data regarding FUS-BBBO-enhanced liquid biopsy for primary brain tumors. A systematic search was conducted in PubMed and Embase databases with key terms "brain tumors", "liquid biopsy", "FUS" and their synonyms, in accordance with PRISMA statement guidelines. Five preclinical and two clinical studies were included. Preclinical studies utilized mouse, rat and porcine glioma models. Biomarker levels were found to be higher in sonicated groups compared to control groups. Both stable and inertial microbubble cavitation increased biomarker levels, whereas only inertial cavitation induced microhemorrhages. In clinical studies involving 14 patients with high-grade brain tumors, biomarker levels were increased after FUS-BBBO with stable cavitation. In conclusion, FUS-BBBO-enhanced liquid biopsy using stable cavitation shows diagnostic potential for primary brain tumors. Further research is imperative before integrating FUS-BBBO for liquid biopsy enhancement into clinical practice.
Collapse
Affiliation(s)
- Akke Bakker
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
- Department of Neurology & Neurosurgery, UMC Utrecht Brain Center, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Anna E. Ixkes
- Biomedical Sciences, Utrecht University, Heidelberglaan 8, 3584 CS Utrecht, The Netherlands
| | - Hema Venugopal
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
- Department of Neurology & Neurosurgery, UMC Utrecht Brain Center, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Mario G. Ries
- Imaging Division, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Nathalie S. M. Lak
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Filip Y. F. L. de Vos
- Department of Medical Oncology, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Dannis G. van Vuurden
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Tom J. Snijders
- Department of Neurology & Neurosurgery, UMC Utrecht Brain Center, UMC Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
35
|
Habashy KJ, Dmello C, Chen L, Arrieta VA, Kim KS, Gould A, Youngblood MW, Bouchoux G, Burdett KB, Zhang H, Canney M, Stupp R, Sonabend AM. Paclitaxel and Carboplatin in Combination with Low-intensity Pulsed Ultrasound for Glioblastoma. Clin Cancer Res 2024; 30:1619-1629. [PMID: 38295144 DOI: 10.1158/1078-0432.ccr-23-2367] [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: 08/05/2023] [Revised: 11/22/2023] [Accepted: 01/29/2024] [Indexed: 02/02/2024]
Abstract
PURPOSE We recently reported on clinical trials for patients with recurrent glioblastoma where low-intensity pulsed ultrasound and microbubbles (LIPU/MB) improved paclitaxel or carboplatin delivery into the brain. Here, we report variable local tumor control with paclitaxel at the maximal/target dose in our phase I trial (NCT04528680). To address this, we investigated the combination of paclitaxel with carboplatin in preclinical glioma models. EXPERIMENTAL DESIGN We performed MRI-based analysis to evaluate disease control in patients from our trial. We studied the cytotoxicity of paclitaxel and carboplatin against 11 human glioma lines as monotherapy and in combination at concentrations derived from human intraoperative studies. Synergy was assessed with the Loewe model and the survival benefit evaluated in two xenografts. We examined the effects on cell cycle progression, DNA damage, and apoptosis. RESULTS Patients treated with paclitaxel and LIPU/MB exhibited variable local tumor control, which correlated with overall survival. We observed limited cross-resistance to paclitaxel and carboplatin in glioma lines, with almost a third of them being exclusively susceptible to one drug. This combination led to susceptibility of 81% of lines and synergy in 55% of them. The combination proved more efficacious in two intracranial xenografts when administered with LIPU/MB, leading to complementary effects on cell cycle arrest. CONCLUSIONS Combining paclitaxel and carboplatin in gliomas may be more efficacious than monotherapy, as in other cancers, due to synergy and independent susceptibility to each drug. These results form the basis for an ongoing phase II trial (NCT04528680) where we investigate this combination with LIPU/MB.
Collapse
Affiliation(s)
- Karl J Habashy
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Crismita Dmello
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Li Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Victor A Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kwang-Soo Kim
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Andrew Gould
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Mark W Youngblood
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Kirsten B Burdett
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hui Zhang
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Roger Stupp
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
36
|
Li Y, Zhao Q, Zhu X, Zhou L, Song P, Liu B, Tian D, Chen Q, Zhou J, Deng G. Self-Assembled nanoparticles of natural bioactive molecules enhance the delivery and efficacy of paclitaxel in glioblastoma. CNS Neurosci Ther 2024; 30:e14528. [PMID: 38044793 PMCID: PMC11017454 DOI: 10.1111/cns.14528] [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: 08/20/2023] [Revised: 10/28/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common primary malignant tumor in the central nervous system. Paclitaxel (PTX) is a well-established and highly effective anti-cancer drug for peripheral solid tumors. However, the application of PTX in GBM is hindered by several limitations, including poor water solubility, restricted entry across the blood-brain barrier (BBB), and enhanced excretion by efflux transporters. P-glycoprotein (P-gp) is a crucial efflux transporter that is abundantly present in cerebral vascular endothelial cells and GBM cells. It plays a significant role in the exocytosis of PTX within tumor tissues. METHODS Recently, we have developed a novel technique for creating self-assembled nanoparticles utilizing a range of natural bioactive molecules. These nanoparticles can encapsulate insoluble drugs and effectively cross the BBB. In additional, we revealed that certain nanoparticles have the potential to act as P-gp inhibitors, thereby reducing the excretion of PTX. In this study, we conducted a screening of bioactive molecular nanoparticles to identify those that effectively inhibit the function of P-gp transporters. RESULTS Among the candidates, we identified ursolic acid nanoparticles (UA NPs) as the P-gp inhibitors. Furthermore, we prepared co-assembled UA NPs embedded with paclitaxel, referred to as UA-PTX NPs. Our results demonstrate that UA-PTX NPs can enhance the blood concentration of PTX, facilitate its entry into the BBB, and inhibit the function of P-gp, resulting in a decrease in the excretion of PTX. This discovery effectively addressed the above three issues associated with the use of PTX in glioma treatment. CONCLUSIONS UA-PTX NPs demonstrate strong anti-tumor effects and show great potential for treating GBM.
Collapse
Affiliation(s)
- Yong Li
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Qingyu Zhao
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Xinyi Zhu
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Long Zhou
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Ping Song
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Baohui Liu
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Daofeng Tian
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Qianxue Chen
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Jiangbing Zhou
- Department of NeurosurgeryYale UniversityNew HavenConnecticutUSA
| | - Gang Deng
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| |
Collapse
|
37
|
Li J, Long Q, Ding H, Wang Y, Luo D, Li Z, Zhang W. Progress in the Treatment of Central Nervous System Diseases Based on Nanosized Traditional Chinese Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308677. [PMID: 38419366 PMCID: PMC11040388 DOI: 10.1002/advs.202308677] [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: 11/13/2023] [Revised: 02/07/2024] [Indexed: 03/02/2024]
Abstract
Traditional Chinese Medicine (TCM) is widely used in clinical practice to treat diseases related to central nervous system (CNS) damage. However, the blood-brain barrier (BBB) constitutes a significant impediment to the effective delivery of TCM, thus substantially diminishing its efficacy. Advances in nanotechnology and its applications in TCM (also known as nano-TCM) can deliver active ingredients or components of TCM across the BBB to the targeted brain region. This review provides an overview of the physiological and pathological mechanisms of the BBB and systematically classifies the common TCM used to treat CNS diseases and types of nanocarriers that effectively deliver TCM to the brain. Additionally, drug delivery strategies for nano-TCMs that utilize in vivo physiological properties or in vitro devices to bypass or cross the BBB are discussed. This review further focuses on the application of nano-TCMs in the treatment of various CNS diseases. Finally, this article anticipates a design strategy for nano-TCMs with higher delivery efficiency and probes their application potential in treating a wider range of CNS diseases.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
| | - Qingyin Long
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
| | - Huang Ding
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
| | - Yang Wang
- Institute of Integrative MedicineDepartment of Integrated Traditional Chinese and Western MedicineXiangya HospitalCentral South University ChangshaChangsha410008China
| | - Dan Luo
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
| | - Zhou Li
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
| | - Wei Zhang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
| |
Collapse
|
38
|
Tazhibi M, McQuillan N, Wei HJ, Gallitto M, Bendau E, Webster Carrion A, Berg X, Kokossis D, Zhang X, Zhang Z, Jan CI, Mintz A, Gartrell RD, Syed HR, Fonseca A, Pavisic J, Szalontay L, Konofagou EE, Zacharoulis S, Wu CC. Focused ultrasound-mediated blood-brain barrier opening is safe and feasible with moderately hypofractionated radiotherapy for brainstem diffuse midline glioma. J Transl Med 2024; 22:320. [PMID: 38555449 PMCID: PMC10981822 DOI: 10.1186/s12967-024-05096-9] [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: 03/22/2023] [Accepted: 03/15/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Diffuse midline glioma (DMG) is a pediatric tumor with dismal prognosis. Systemic strategies have been unsuccessful and radiotherapy (RT) remains the standard-of-care. A central impediment to treatment is the blood-brain barrier (BBB), which precludes drug delivery to the central nervous system (CNS). Focused ultrasound (FUS) with microbubbles can transiently and non-invasively disrupt the BBB to enhance drug delivery. This study aimed to determine the feasibility of brainstem FUS in combination with clinical doses of RT. We hypothesized that FUS-mediated BBB-opening (BBBO) is safe and feasible with 39 Gy RT. METHODS To establish a safety timeline, we administered FUS to the brainstem of non-tumor bearing mice concurrent with or adjuvant to RT; our findings were validated in a syngeneic brainstem murine model of DMG receiving repeated sonication concurrent with RT. The brainstems of male B6 (Cg)-Tyrc-2J/J albino mice were intracranially injected with mouse DMG cells (PDGFB+, H3.3K27M, p53-/-). A clinical RT dose of 39 Gy in 13 fractions (39 Gy/13fx) was delivered using the Small Animal Radiation Research Platform (SARRP) or XRAD-320 irradiator. FUS was administered via a 0.5 MHz transducer, with BBBO and tumor volume monitored by magnetic resonance imaging (MRI). RESULTS FUS-mediated BBBO did not affect cardiorespiratory rate, motor function, or tissue integrity in non-tumor bearing mice receiving RT. Tumor-bearing mice tolerated repeated brainstem BBBO concurrent with RT. 39 Gy/13fx offered local control, though disease progression occurred 3-4 weeks post-RT. CONCLUSION Repeated FUS-mediated BBBO is safe and feasible concurrent with RT. In our syngeneic DMG murine model, progression occurs, serving as an ideal model for future combination testing with RT and FUS-mediated drug delivery.
Collapse
Affiliation(s)
- Masih Tazhibi
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Nicholas McQuillan
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Hong-Jian Wei
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Matthew Gallitto
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Ethan Bendau
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Andrea Webster Carrion
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Xander Berg
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Danae Kokossis
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA
| | - Xu Zhang
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Zhiguo Zhang
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Chia-Ing Jan
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
| | - Akiva Mintz
- Department of Radiology, Columbia University, New York, NY, 10027, USA
| | - Robyn D Gartrell
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Hasan R Syed
- Department of Neurosurgery, Children's National Hospital, Washington, DC, USA
- George Washington University, Washington, DC, USA
| | - Adriana Fonseca
- George Washington University, Washington, DC, USA
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, DC, USA
- The Brain Tumor Institute, Children's National Hospital, Washington, DC, USA
| | - Jovana Pavisic
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| | - Luca Szalontay
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Stergios Zacharoulis
- Division of Pediatric Hematology Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY, 10032, USA.
- Bristol Myers Squibb, Princeton, NJ, 08901, USA.
| | - Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University Irving Medical Center, 622 W. 168th Street, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, New York, NY, 10032, USA.
| |
Collapse
|
39
|
Xie Z, Ye J, Gao X, Chen H, Chen M, Lian J, Ma J, Wang H. Evaluation of nanoparticle albumin-bound paclitaxel loaded macrophages for glioblastoma treatment based on a microfluidic chip. Front Bioeng Biotechnol 2024; 12:1361682. [PMID: 38562665 PMCID: PMC10982336 DOI: 10.3389/fbioe.2024.1361682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction: Glioblastoma (GBM) is a primary brain malignancy with a dismal prognosis and remains incurable at present. In this study, macrophages (MΦ) were developed to carry nanoparticle albumin-bound paclitaxel (nab-PTX) to form nab-PTX/MΦ. The aim of this study is to use a GBM-on-a-chip to evaluate the anti-GBM effects of nab-PTX/MΦ. Methods: In this study, we constructed nab-PTX/MΦ by incubating live MΦ with nab-PTX. We developed a microfluidic chip to co-culture GBM cells and human umbilical vein endothelial cells, mimicking the simplified blood-brain barrier and GBM. Using a syringe pump, we perform sustainable perfusion of nutrient media. To evaluate the anti-GBM effects nab-PTX/MΦ, we treated the GBM-on-a-chip model with nab-PTX/MΦ and investigated GBM cell proliferation, migration, and spheroid formation. Results: At the chosen concentration, nab-PTX did not significantly affect the viability, chemotaxis and migration of MΦ. The uptake of nab-PTX by MΦ occurred within 1 h of incubation and almost reached saturation at 6 h. Additionally, nab-PTX/MΦ exhibited the M1 phenotype, which inhibits tumor progression. Following phagocytosis, MΦ were able to release nab-PTX, and the release of nab-PTX by MΦ had nearly reached its limit at 48 h. Compared with control group and blank MΦ group, individual nab-PTX group and nab-PTX/MΦ group could inhibit tumor proliferation, invasion and spheroid formation. Meanwhile, the anti-GBM effect of nab-PTX/MΦ was more significant than nab-PTX. Discussion: Our findings demonstrate that nab-PTX/MΦ has a significant anti-GBM effect compared to individual nab-PTX or MΦ administration, suggesting MΦ as potential drug delivery vectors for GBM therapy. Furthermore, the developed GBM-on-a-chip model provides a potential ex vivo platform for innovative cell-based therapies and tailored therapeutic strategies for GBM.
Collapse
Affiliation(s)
- Zuorun Xie
- Department of Neurosurgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Junyi Ye
- Department of Neurosurgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai, China
| | - Hang Chen
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Maosong Chen
- Department of Neurosurgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jiangfang Lian
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Jingyun Ma
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Hongcai Wang
- Department of Neurosurgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
40
|
Villa A, De Mitri Z, Vincenti S, Crippa E, Castiglioni L, Gelosa P, Rebecchi M, Tosi D, Brunialti E, Oevermann A, Falleni M, Sironi L, Bello L, Mazzaferro V, Ciana P. Canine glioblastoma-derived extracellular vesicles as precise carriers for glioblastoma imaging: Targeting across the blood-brain barrier. Biomed Pharmacother 2024; 172:116201. [PMID: 38306846 DOI: 10.1016/j.biopha.2024.116201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/04/2024] Open
Abstract
The treatment of glioblastoma (GBM) faces significant challenges due to the difficulty of delivering drugs through the blood-brain barrier (BBB). Extracellular vesicles (EVs) have emerged as potential carriers for targeted drug delivery to brain tumors. However, their use and distribution in the presence of an intact BBB and their ability to target GBM tissue are still under investigation. This study explored the use of EVs for GBM targeting across the BBB. Canine plasma EVs from healthy dogs and dogs with glioma were isolated, characterized, and loaded with diagnostic agents. Biodistribution studies were conducted in healthy murine models and a novel intranasal model that preserved BBB integrity while initiating early-stage GBM growth. This model assessed EVs' potential for delivering the contrast agent gadoteric acid to intracranial tumors. Imaging techniques, such as bioluminescence and MRI, confirmed EVs' targeting and delivery capabilities thus revealing a selective accumulation of canine glioma-derived EVs in brain tissue under physiological conditions. In the model of brain tumor, MRI experiments demonstrated the ability of EVs to accumulate gadoteric acid within GBM to enhance contrast of the tumoral mass, even when BBB integrity is maintained. This study underscores the potential of EVs derived from glioma for the targeted delivery of drugs to glioblastoma. EVs from dogs with glioma showed capacity to traverse the BBB and selectively accumulate within the brain tumor. Overall, this research represents a foundation for the application of autologous EVs to precision glioblastoma treatment, addressing the challenge of BBB penetration and targeting specificity in brain cancer therapy.
Collapse
Affiliation(s)
- Alessandro Villa
- Department of Health Sciences, University of Milan, via A. di Rudinì, 8, 20142, Milano, Italy
| | - Zemira De Mitri
- Department of Health Sciences, University of Milan, via A. di Rudinì, 8, 20142, Milano, Italy
| | - Simona Vincenti
- Department of Clinical Veterinary Medicine, Vetsuisse Faculty, University of Bern, Länggassstrasse 124, 3001 Bern, Switzerland
| | - Elisabetta Crippa
- Department of Health Sciences, University of Milan, via A. di Rudinì, 8, 20142, Milano, Italy
| | - Laura Castiglioni
- Department of Pharmaceutical Sciences, University of Milan, via Balzaretti, 20133 Milano, Italy
| | - Paolo Gelosa
- Department of Pharmaceutical Sciences, University of Milan, via Balzaretti, 20133 Milano, Italy
| | - Monica Rebecchi
- Department of Health Sciences, University of Milan, via A. di Rudinì, 8, 20142, Milano, Italy
| | - Delfina Tosi
- Department of Health Sciences, University of Milan, via A. di Rudinì, 8, 20142, Milano, Italy
| | - Electra Brunialti
- Department of Health Sciences, University of Milan, via A. di Rudinì, 8, 20142, Milano, Italy
| | - Anna Oevermann
- Division of Neurological Sciences, Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Länggassstrasse 124, 3001 Bern, Switzerland
| | - Monica Falleni
- Department of Health Sciences, University of Milan, via A. di Rudinì, 8, 20142, Milano, Italy
| | - Luigi Sironi
- Department of Pharmaceutical Sciences, University of Milan, via Balzaretti, 20133 Milano, Italy
| | - Lorenzo Bello
- Department of Oncology and Hemato-Oncology, Università Degli Studi di Milano, Via Festa del Perdono 7, 20122 Milano, Italy
| | - Vincenzo Mazzaferro
- Department of Oncology and Hemato-Oncology, Università Degli Studi di Milano, Via Festa del Perdono 7, 20122 Milano, Italy; HPB Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale Tumori (INT), Via Giacomo Venezian, 1, 20133 Milano, Italy
| | - Paolo Ciana
- Department of Health Sciences, University of Milan, via A. di Rudinì, 8, 20142, Milano, Italy.
| |
Collapse
|
41
|
Moscona-Nissan A, Habashy KJ, Arrieta VA, Sonabend AM, Dmello C. Combining causal and correlative approaches to discover biomarkers of response to paclitaxel. Oncotarget 2024; 15:117-122. [PMID: 38329732 PMCID: PMC10852058 DOI: 10.18632/oncotarget.28549] [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: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 02/09/2024] Open
Abstract
We recently discovered a putative paclitaxel response predictive biomarker for glioblastoma and breast cancer using the whole genome CRISPR knockout screen. The biomarker candidate was validated in two independent breast cancer patient cohorts that received taxane treatment. To further evaluate the potential application of this biomarker in the clinic for patients with glioblastoma, a prospective validation in cohorts of patients with glioblastoma is essential and will be performed as part of our ongoing phase II clinical trial (NCT04528680). The validation of novel biomarkers of susceptibility to therapy is critical to elucidate the efficacy signal of therapeutic agents. This is especially important in the context of glioblastoma, where therapeutic benefit is variable and unpredictable, leading to negative trials, yet the outcome of subset of patients has outperformed expectations.
Collapse
Affiliation(s)
| | - Karl J. Habashy
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago IL 60611, USA
- Northwestern Medicine, Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL 60611, USA
| | - Victor A. Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago IL 60611, USA
- Northwestern Medicine, Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL 60611, USA
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Adam M. Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago IL 60611, USA
- Northwestern Medicine, Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL 60611, USA
| | - Crismita Dmello
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago IL 60611, USA
- Northwestern Medicine, Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL 60611, USA
| |
Collapse
|
42
|
Miranda O, Fan P, Qi X, Wang H, Brannock MD, Kosten T, Ryan ND, Kirisci L, Wang L. Prediction of adverse events risk in patients with comorbid post-traumatic stress disorder and alcohol use disorder using electronic medical records by deep learning models. Drug Alcohol Depend 2024; 255:111066. [PMID: 38217979 PMCID: PMC10853953 DOI: 10.1016/j.drugalcdep.2023.111066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND Identifying co-occurring mental disorders and elevated risk is vital for optimization of healthcare processes. In this study, we will use DeepBiomarker2, an updated version of our deep learning model to predict the adverse events among patients with comorbid post-traumatic stress disorder (PTSD) and alcohol use disorder (AUD), a high-risk population. METHODS We analyzed electronic medical records of 5565 patients from University of Pittsburgh Medical Center to predict adverse events (opioid use disorder, suicide related events, depression, and death) within 3 months at any encounter after the diagnosis of PTSD+AUD by using DeepBiomarker2. We integrated multimodal information including: lab tests, medications, co-morbidities, individual and neighborhood level social determinants of health (SDoH), psychotherapy and veteran data. RESULTS DeepBiomarker2 achieved an area under the receiver operator curve (AUROC) of 0.94 on the prediction of adverse events among those PTSD+AUD patients. Medications such as vilazodone, dronabinol, tenofovir, suvorexant, modafinil, and lamivudine showed potential for risk reduction. SDoH parameters such as cognitive behavioral therapy and trauma focused psychotherapy lowered risk while active veteran status, income segregation, limited access to parks and greenery, low Gini index, limited English-speaking capacity, and younger patients increased risk. CONCLUSIONS Our improved version of DeepBiomarker2 demonstrated its capability of predicting multiple adverse event risk with high accuracy and identifying potential risk and beneficial factors.
Collapse
Affiliation(s)
- Oshin Miranda
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences/School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Peihao Fan
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences/School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiguang Qi
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences/School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Haohan Wang
- School of Information Sciences at the University of Illinois Urbana-Champaign, Champaign, IL 61820, USA
| | | | - Thomas Kosten
- Menninger Department of Psychiatry, Baylor College of Medicine, Houston, TX 77030, USA
| | - Neal David Ryan
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Levent Kirisci
- University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15213, USA
| | - LiRong Wang
- Computational Chemical Genomics Screening Center, Department of Pharmaceutical Sciences/School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
43
|
Pandey S, Cholak ME, Yadali R, Sosman JA, Tetreault MP, Fang D, Pollack SM, Gnjatic S, Obeng RC, Lyerly HK, Sonabend AM, Guevara-Patiño JA, Butterfield LH, Zhang B, Maecker HT, Le Poole IC. Immune Assessment Today: Optimizing and Standardizing Efforts to Monitor Immune Responses in Cancer and Beyond. Cancers (Basel) 2024; 16:475. [PMID: 38339227 PMCID: PMC10854499 DOI: 10.3390/cancers16030475] [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: 12/09/2023] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
As part of a symposium, current and former directors of Immune Monitoring cores and investigative oncologists presented insights into the past, present and future of immune assessment. Dr. Gnjatic presented a classification of immune monitoring technologies ranging from universally applicable to experimental protocols, while emphasizing the need for assay harmonization. Dr. Obeng discussed physiologic differences among CD8 T cells that align with anti-tumor responses. Dr. Lyerly presented the Soldano Ferrone lecture, commemorating the passionate tumor immunologist who inspired many, and covered a timeline of monitoring technology development and its importance to immuno-oncology. Dr. Sonabend presented recent achievements in glioblastoma treatment, accentuating the range of monitoring techniques that allowed him to refine patient selection for clinical trials. Dr. Guevara-Patiño focused on hypoxia within the tumor environment and stressed that T cell viability is not to be confused with functionality. Dr. Butterfield accentuated monitoring of dendritic cell metabolic (dys)function as a determinant for tumor vaccine success. Lectures were interspersed with select abstract presentations. To summarize the concepts, Dr. Maecker from Stanford led an informative forum discussion, pointing towards the future of immune monitoring. Immune monitoring continues to be a guiding light towards effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Surya Pandey
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
| | - Meghan E. Cholak
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
| | - Rishita Yadali
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
| | - Jeffrey A. Sosman
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Marie-Pier Tetreault
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Deyu Fang
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Seth M. Pollack
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Sacha Gnjatic
- Human Immune Monitoring Center, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Rebecca C. Obeng
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - H. Kim Lyerly
- Center for Applied Therapeutics, Duke Cancer Center, Duke University, Durham, NC 27710, USA;
| | - Adam M. Sonabend
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | | | - Lisa H. Butterfield
- Merck Research Laboratories, Boston, MA 02115, USA;
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
| | - Bin Zhang
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| | - Holden T. Maecker
- Human Immune Monitoring Center, Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - I. Caroline Le Poole
- Immunotherapy Assessment Core, Chicago, IL 60611, USA; (S.P.); (M.E.C.); (R.Y.); (B.Z.)
- Lurie Comprehensive Cancer Center, Northwestern University at Chicago, Chicago, IL 60611, USA; (J.A.S.); (M.-P.T.); (D.F.); (S.M.P.); (A.M.S.)
| |
Collapse
|
44
|
Ahmed M, Semreen AM, Giddey AD, Ramadan WS, El-Awady R, Soares NC, El-Huneidi W, Bustanji Y, Alqudah MAY, Alzoubi KH, Semreen MH. Proteomic and metabolomic signatures of U87 glioblastoma cells treated with cisplatin and/or paclitaxel. Ann Med 2024; 55:2305308. [PMID: 38253025 PMCID: PMC10810643 DOI: 10.1080/07853890.2024.2305308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a primary malignancy of the central nervous system and is classified as a grade IV astrocytoma by the World Health Organization (WHO). Although GBM rarely metastasizes, its prognosis remains poor. Moreover, the standard treatment for GBM, temozolomide (TMZ), is associated with chemoresistance, which is a major factor behind GBM-related deaths. Investigating drugs with repurposing potential in the context of GBM is worthwhile to bypass lengthy bench-to-bedside research. The field of omics has garnered significant interest in scientific research because of its potential to delineate the intricate regulatory network underlying tumor development. In particular, proteomic and metabolomic analyses are powerful approaches for the investigation of metabolic enzymes and intermediate metabolites since they represent the functional end of the cancer phenotype. METHODS We chose two of the most widely prescribed anticancer drugs, cisplatin and paclitaxel. To our knowledge, the current literature lacks studies examining their effects on metabolic and proteomic alterations in GBM. We employed the mass spectrometry technological platform 'UHPLC-Q-TOF-MS/MS' to examine the changes in the proteome and metabolome profiles of the U87 cell line with defined concentrations of cisplatin and/or paclitaxel via an untargeted approach. RESULTS A total of 1,419 distinct proteins and 90 metabolites were generated, and subsequent analysis was performed. We observed that upon treatment with cisplatin (9.5 μM), U87 cells exhibited apparent efforts to cope with this exogenous stressor, understanding the effect of paclitaxel (5.3 μM) on altering the transport machinery of the cell, and how the combination of cisplatin and/or paclitaxel suggests potential interactions with promising benefits in GBM therapeutics. CONCLUSION Our research provides a detailed map of alterations in response to cisplatin and paclitaxel treatment, provides crucial insights into the molecular basis of their action, and paves the way for further research to identify molecular targets for this elusive malignancy.
Collapse
Affiliation(s)
- Munazza Ahmed
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ahlam M. Semreen
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Alexander D. Giddey
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Wafaa S. Ramadan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Raafat El-Awady
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Nelson C. Soares
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Lisbon, Portugal
| | - Waseem El-Huneidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Yasser Bustanji
- Department of Basic and Clinical Pharmacology, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Mohammad A. Y. Alqudah
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H. Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
45
|
Farooq M, Scalia G, Umana GE, Parekh UA, Naeem F, Abid SF, Khan MH, Zahra SG, Sarkar HP, Chaurasia B. A Systematic Review of Nanomedicine in Glioblastoma Treatment: Clinical Efficacy, Safety, and Future Directions. Brain Sci 2023; 13:1727. [PMID: 38137175 PMCID: PMC10742051 DOI: 10.3390/brainsci13121727] [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: 11/02/2023] [Revised: 12/13/2023] [Accepted: 12/17/2023] [Indexed: 12/24/2023] Open
Abstract
(1) Background: Glioblastoma (GBM) is categorized as a grade IV astrocytoma by the World Health Organization (WHO), representing the most aggressive and prevalent form of glioma. It presents a significant clinical challenge, with limited treatment options and poor prognosis. This systematic review evaluates the efficacy and safety of various nanotherapy approaches for GBM and explores future directions in tumor management. Nanomedicine, which involves nanoparticles in the 1-100 nm range, shows promise in improving drug delivery and targeting tumor cells. (2) Methods: Following PRISMA guidelines, a systematic search of databases including Google Scholar, NCBI PubMed, Cochrane Library, and ClinicalTrials.gov was conducted to identify clinical trials on GBM and nanomedicine. The primary outcome measures were median overall survival, progression-free survival, and quality of life assessed through Karnofsky performance scores. The safety profile was assessed by adverse events. (3) Results: The analysis included 225 GBM patients, divided into primary and recurrent sub-populations. Primary GBM patients had a median overall survival of 6.75 months, while recurrent GBM patients had a median overall survival of 9.7 months. The mean PFS period was 2.3 months and 3.92 months in primary GBM and recurrent GBM patients, respectively. Nanotherapy showed an improvement in quality of life, with KPS scores increasing after treatment in recurrent GBM patients. Adverse events were observed in 14.2% of patients. Notably, Bevacizumab therapy exhibited better survival outcomes but with a higher incidence of adverse events. (4) Conclusions: Nanotherapy offers a modest increase in survival with fewer severe side effects. It shows promise in improving the quality of life, especially in recurrent GBM patients. However, it falls short in terms of overall survival compared to Bevacizumab. The heterogeneous nature of treatment protocols and reporting methods highlights the need for standardized multicenter trials to further evaluate the potential of nanomedicine in GBM management.
Collapse
Affiliation(s)
- Minaam Farooq
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10021, USA;
| | - Gianluca Scalia
- Neurosurgery Unit, Department of Head and Neck Surgery, Garibaldi Hospital, 95123 Catania, Italy
| | - Giuseppe E. Umana
- Department of Neurosurgery, Gamma Knife and Trauma Center, Cannizzaro Hospital, 95126 Catania, Italy;
| | - Urja A. Parekh
- German Cancer Research Center, 69120 Heidelberg, Germany;
| | - Faiza Naeem
- Department of Neurosurgery, King Edward Medical University, Lahore 54000, Pakistan; (F.N.); (S.F.A.); (M.H.K.); (S.G.Z.)
| | - Sayeda Fatima Abid
- Department of Neurosurgery, King Edward Medical University, Lahore 54000, Pakistan; (F.N.); (S.F.A.); (M.H.K.); (S.G.Z.)
| | - Muhammad Hammad Khan
- Department of Neurosurgery, King Edward Medical University, Lahore 54000, Pakistan; (F.N.); (S.F.A.); (M.H.K.); (S.G.Z.)
| | - Shah Gul Zahra
- Department of Neurosurgery, King Edward Medical University, Lahore 54000, Pakistan; (F.N.); (S.F.A.); (M.H.K.); (S.G.Z.)
| | - Hrishikesh P. Sarkar
- Department of Neurological Sciences, Kokilaben Dhirubhai Ambani Hospital, Mumbai 400053, India;
| | - Bipin Chaurasia
- Department of Neurosurgery, Neurosurgery Clinic, Birgunj 44300, Nepal;
| |
Collapse
|
46
|
Lin Y, Gan L, Ren L, Ma C, Dai M, Qian K, Ye Q, Lin X. Acupuncture with specific mode electro-stimulation effectively and transiently opens the BBB through Shh signaling pathway. Neuroreport 2023; 34:873-886. [PMID: 37942738 DOI: 10.1097/wnr.0000000000001970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
To explore a new method that patients with brain diseases such as stroke sequelae are hindered by blood-brain barrier (BBB) in clinical treatment. Research preliminarily found that acupuncture with specific mode electro-stimulation (EA) to open BBB-assisted drug delivery may be is an effective means to improve the clinical efficacy of brain disease patients. So here we further explore the features and mechanism. Middle cerebral artery occlusion/R recovery rats were employed as the animal model. Laser Doppler monitoring cerebral blood flow decreased to 45 ± 10% of the baseline value as modeling criteria and TTC staining observed infarcted areas of brain tissue. The permeability of FITC-Dextran and EB in the frontal lobe of rats was observed by microscope. After that, Western blot and Immunofluorescence staining for the detection of the shh and Gli1 signal molecule, Claudin-5 Occludin ZO-1 tight junction (TJ) proteins. EA can open the BBB stably and effectively, and has the characteristics of starting to close soon after the end of EA; EA inhibits the Shh-Gli1 signaling pathway, and downregulates Occludin ZO-1 TJ proteins. These results suggest that EA is safe and reversible in opening the BBB, and its mechanism is related to the inhibition of Shh signaling pathway to down-regulate the expression of TJ proteins.
Collapse
Affiliation(s)
- Yubo Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Ahmed MH, Canney M, Carpentier A, Thanou M, Idbaih A. Unveiling the enigma of the blood-brain barrier in glioblastoma: current advances from preclinical and clinical studies. Curr Opin Oncol 2023; 35:522-528. [PMID: 37681417 PMCID: PMC10566587 DOI: 10.1097/cco.0000000000000990] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
PURPOSE OF REVIEW Glioblastoma (GBM), the most prevalent primary brain malignancy in adults, poses significant challenges in terms of treatment. Current therapeutic strategies for GBM patients involve maximal safe resection, followed by radiotherapy with concurrent and adjuvant temozolomide. However, despite this multimodal approach for GBM, the prognosis of GBM patients remains dismal because of their inherent primary and secondary resistances to treatments. RECENT FINDINGS Several molecular and cellular mechanisms, including the presence of the blood-brain barrier (BBB), contribute to these resistances. The BBB, comprising multiple layers surrounding brain vessels, acts as a barrier limiting effective drug delivery to the brain. Invasive and noninvasive tools to deliver drugs and pharmaceutical formulations locally or systemically are continuously evolving to overcome the BBB in GBM toward improving drug bioavailability in the brain and reducing systemic toxicities. SUMMARY Preliminary studies utilizing these approaches have demonstrated promising results in terms of safety and signals of efficacy during early-phase clinical trials. However, further work through additional clinical trials is necessary to evaluate the potential clinical benefits for GBM patients.
Collapse
Affiliation(s)
- Mohammed H. Ahmed
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | | | - Alexandre Carpentier
- Sorbonne Université, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurochirurgie
| | - Maya Thanou
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Ahmed Idbaih
- Sorbonne Université, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, DMU Neurosciences, Service de Neurologie 2-Mazarin, Paris, France
| |
Collapse
|
48
|
Qu S, Qi S, Zhang H, Li Z, Wang K, Zhu T, Ye R, Zhang W, Huang G, Yi GZ. Albumin-bound paclitaxel augment temozolomide treatment sensitivity of glioblastoma cells by disrupting DNA damage repair and promoting ferroptosis. J Exp Clin Cancer Res 2023; 42:285. [PMID: 37891669 PMCID: PMC10612313 DOI: 10.1186/s13046-023-02843-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Temozolomide (TMZ) treatment efficacy in glioblastoma (GBM) patients has been limited by resistance in the clinic. Currently, there are no clinically proven therapeutic options available to restore TMZ treatment sensitivity. Here, we investigated the potential of albumin-bound paclitaxel (ABX), a novel microtubule targeting agent, in sensitizing GBM cells to TMZ and elucidated its underlying molecular mechanism. METHODS A series of in vivo and in vitro experiments based on two GBM cell lines and two primary GBM cells were designed to evaluate the efficacy of ABX in sensitizing GBM cells to TMZ. Further proteomic analysis and validation experiments were performed to explore the underlying molecular mechanism. Finally, the efficacy and mechanism were validated in GBM patients derived organoids (PDOs) models. RESULTS ABX exhibited a synergistic inhibitory effect on GBM cells when combined with TMZ in vitro. Combination treatment of TMZ and ABX was highly effective in suppressing GBM progression and significantly prolonged the survival oforthotopic xenograft nude mice, with negligible side effects. Further proteomic analysis and experimental validation demonstrated that the combined treatment of ABX and TMZ can induce sustained DNA damage by disrupting XPC and ERCC1 expression and nuclear localization. Additionally, the combination treatment can enhance ferroptosis through regulating HOXM1 and GPX4 expression. Preclinical drug-sensitivity testing based on GBM PDOs models confirmed that combination therapy was significantly more effective than conventional TMZ monotherapy. CONCLUSION Our findings suggest that ABX has the potential to enhance TMZ treatment sensitivity in GBM, which provides a promising therapeutic strategy for GBM patients.
Collapse
Affiliation(s)
- Shanqiang Qu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Nanfang Glioma Center, Guangzhou, Guangdong, People's Republic of China
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Nanfang Glioma Center, Guangzhou, Guangdong, People's Republic of China
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Huayang Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Nanfang Glioma Center, Guangzhou, Guangdong, People's Republic of China
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Kaicheng Wang
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Taichen Zhu
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Rongxu Ye
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Wanghao Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
- Nanfang Glioma Center, Guangzhou, Guangdong, People's Republic of China.
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Guo-Zhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
- The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
- Nanfang Glioma Center, Guangzhou, Guangdong, People's Republic of China.
- Institute of Brain Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
49
|
Nie Q, Chen W, Zhang T, Ye S, Ren Z, Zhang P, Wen J. Iron oxide nanoparticles induce ferroptosis via the autophagic pathway by synergistic bundling with paclitaxel. Mol Med Rep 2023; 28:198. [PMID: 37681444 PMCID: PMC10510030 DOI: 10.3892/mmr.2023.13085] [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: 02/09/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023] Open
Abstract
In recent years, inhibiting tumor cell activity by triggering cell ferroptosis has become a research hotspot. The development of generic targeted nanotherapeutics might bring new ideas for non‑invasive applications. Currently, the potential mechanism underlying the universal application of paclitaxel (PTX)‑loaded iron oxide nanoparticles (IONP@PTX) to different types of tumors is unclear. The present study aimed to prepare IONP@PTX for targeted cancer therapy and further explore the potential mechanisms underlying the inhibitory effects of this material on the NCI‑H446 human small cell lung cancer and brain M059K malignant glioblastoma cell lines. First, a CCK‑8 assay was performed to determine cell viability, and then the combination index for evaluating drug combination interaction effect was evaluated. Intracellular reactive oxygen species (ROS) and lipid peroxidation levels were monitored using a DCFH‑DA fluorescent probe and a C11‑BODIPY™ fluorescent probe, respectively. Furthermore, western blotting assay was performed to determine the expression of autophagy‑ and iron death‑related proteins. The experimental results showed that, compared with either IONP monotherapy, PTX monotherapy, or IONP + PTX, IONP@PTX exerted a synergistic effect on the viability of both cell types, with significantly increased total iron ion concentration, ROS levels and lipid peroxidation levels. IONP@PTX significantly increased the expression of autophagy‑related proteins Beclin 1 and histone deacetylase 6 (HDAC6) in both cell lines (P<0.05), increased the expression of light chain 3 (LC3)‑II/I in NCI‑H446 cells (P<0.05) and decreased that of sequestosome1 (p62) in M059K cells (P<0.05). Moreover, the addition of rapamycin enhanced the IONP@PTX‑induced the upregulation of Beclin 1, LC3‑II/I and HDAC6 and the downregulation of mTORC1 protein in both cell lines (P<0.05). Moreover, rapamycin enhanced the IONP@PTX‑induced downregulation of p62 protein in NCI‑H446 cells (P<0.05), suggesting that IONP@PTX induces ferroptosis, most likely through autophagy. Collectively, the present findings show that IONP works synergistically with PTX to induce ferroptosis via the autophagic pathway.
Collapse
Affiliation(s)
- Qi Nie
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi 541104, P.R. China
| | - Wenqing Chen
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi 541104, P.R. China
| | - Tianmei Zhang
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Shangrong Ye
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Zhongyu Ren
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Peng Zhang
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Jian Wen
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi 541104, P.R. China
| |
Collapse
|
50
|
Bérard C, Truillet C, Larrat B, Dhermain F, Estève MA, Correard F, Novell A. Anticancer drug delivery by focused ultrasound-mediated blood-brain/tumor barrier disruption for glioma therapy: From benchside to bedside. Pharmacol Ther 2023; 250:108518. [PMID: 37619931 DOI: 10.1016/j.pharmthera.2023.108518] [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: 07/17/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
The therapeutic management of gliomas remains particularly challenging. Brain tumors present multiple obstacles that make therapeutic innovation complex, mainly due to the presence of blood-tumor and blood-brain barriers (BTB and BBB, respectively) which prevent penetration of anticancer agents into the brain parenchyma. Focused ultrasound-mediated BBB disruption (FUS-BBBD) provides a physical method for non-invasive, local, and reversible BBB disruption. The safety of this technique has been demonstrated in small and large animal models. This approach promises to enhance drug delivery into the brain tumor and therefore to improve survival outcomes by repurposing existing drugs. Several clinical trials continue to be initiated in the last decade. In this review, we provide an overview of the rationale behind the use of FUS-BBBD in gliomas and summarize the preclinical studies investigating different approaches (free drugs, drug-loaded microbubbles and drug-loaded nanocarriers) in combination with this technology in in vivo glioma models. Furthermore, we discuss the current state of clinical trials and devices developed and review the challenges to overcome for clinical use of FUS-BBBD in glioma therapy.
Collapse
Affiliation(s)
- Charlotte Bérard
- Aix Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, Hôpital Timone, Service Pharmacie, 13005 Marseille, France.
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 91401 Orsay, France.
| | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Centre d'études de Saclay, 91191 Gif-sur-Yvette, France.
| | - Frédéric Dhermain
- Radiation Oncology Department, Gustave Roussy University Hospital, 94805 Villejuif, France.
| | - Marie-Anne Estève
- Aix Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, Hôpital Timone, Service Pharmacie, 13005 Marseille, France.
| | - Florian Correard
- Aix Marseille Univ, APHM, CNRS, INP, Inst Neurophysiopathol, Hôpital Timone, Service Pharmacie, 13005 Marseille, France.
| | - Anthony Novell
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Service Hospitalier Frédéric Joliot, 91401 Orsay, France.
| |
Collapse
|