1
|
Gong Z, Zhou D, Wu D, Han Y, Yu H, Shen H, Feng W, Hou L, Chen Y, Xu T. Challenges and material innovations in drug delivery to central nervous system tumors. Biomaterials 2025; 319:123180. [PMID: 39985979 DOI: 10.1016/j.biomaterials.2025.123180] [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: 06/05/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
Central nervous system (CNS) tumors, encompassing a diverse array of neoplasms in the brain and spinal cord, pose significant therapeutic challenges due to their intricate anatomy and the protective presence of the blood-brain barrier (BBB). The primary treatment obstacle is the effective delivery of therapeutics to the tumor site, which is hindered by multiple physiological, biological, and technical barriers, including the BBB. This comprehensive review highlights recent advancements in material science and nanotechnology aimed at surmounting these delivery challenges, with a focus on the development and application of nanomaterials. Nanomaterials emerge as potent tools in designing innovative drug delivery systems that demonstrate the potential to overcome the limitations posed by CNS tumors. The review delves into various strategies, including the use of lipid nanoparticles, polymeric nanoparticles, and inorganic nanoparticles, all of which are engineered to enhance drug stability, BBB penetration, and targeted tumor delivery. Additionally, this review highlights the burgeoning role of theranostic nanoparticles, integrating therapeutic and diagnostic functionalities to optimize treatment efficacy. The exploration extends to biocompatible materials like biodegradable polymers, liposomes, and advanced material-integrated delivery systems such as implantable drug-eluting devices and microfabricated devices. Despite promising preclinical results, the translation of these material-based strategies into clinical practice necessitates further research and optimization.
Collapse
Affiliation(s)
- Zhenyu Gong
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China; Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Dairan Zhou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, 230601, PR China
| | - Yaguang Han
- Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Hao Yu
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, PR China
| | - Haotian Shen
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Lijun Hou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| | - Tao Xu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| |
Collapse
|
2
|
Lee SH, Hofstede RP, Noriega de la Colina A, Gunton JH, Bernstock JD, Traverso G. Implantable systems for neurological chronotherapy. Adv Drug Deliv Rev 2025; 221:115574. [PMID: 40187646 DOI: 10.1016/j.addr.2025.115574] [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: 12/01/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
Implantable systems for neurological chronotherapy are poised to revolutionize the treatment of central nervous system diseases and disorders. These devices enable precise, time-controlled drug delivery aligned with the body's circadian rhythms, optimizing therapeutic outcomes. By bypassing the blood-brain barrier, they achieve high local drug concentrations while minimizing systemic side effects, offering significant advantages for conditions where traditional therapies often fall short. Platforms like SynchroMed II and CraniUS showcase this innovation, providing programmable delivery for conditions such as epilepsy and glioblastoma, with customizable profiles ranging from continuous infusion to timed bolus administration. Preclinical and clinical studies underscore the efficacy of aligning drug delivery with circadian rhythms, enhancing outcomes in chrono-chemotherapy and anti-epileptic treatments. Despite their promise, challenges remain, including the invasiveness of implantation within the brain, device longevity, synchronization complexities, and cost(s). Accordingly, this review explores the current state of implantable neurological systems that may be leveraged for chronotherapy, their applications, limitations, and potential to transform neurological disease/disorder management.
Collapse
Affiliation(s)
- Seung Ho Lee
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Roemer Pott Hofstede
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - John H Gunton
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua D Bernstock
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
3
|
Ahn M, Na Y, Choi H, Lee S, Lee J, Park SA, Jeun SS, Na K. Photoimmuno-Lure Nanoplatform for Enhancing T Cell Expansion in Glioblastoma via Synergistic Treatment of Photodynamic Therapy and Immune Checkpoint Inhibition. Adv Healthc Mater 2025:e2500880. [PMID: 40395101 DOI: 10.1002/adhm.202500880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 05/07/2025] [Indexed: 05/22/2025]
Abstract
The immunosuppressive tumor microenvironment (TME) of glioblastoma (GBM) limits the efficacy of immune checkpoint inhibitors (ICI), primarily due to the absence of cytotoxic T (Tc) cells. In this study, a photoimmuno-lure nanoplatform is presented that combines amphiphilic photosensitizers (PSs) with Atezolizumab leading to the modulation of the TME of GBM and improvement of the therapeutic efficacy through synergistic photodynamic therapy (PDT). The amphiphilic PSs exhibited four-fold higher GBM specificity, superior photostability, and enhanced singlet oxygen generation efficiency (1O2ΦΔ: 0.92) compared to conventional PSs. In in vitro GBM cell lines, amphiphilic PSs increased immune activation cytokines and improved ICI responsiveness compared to single ICI treatment. In addition, similar results are acquired in a GBM 3D spheroid model, showing significantly elevated Tc cell activation. In orthotopic in vivo GBM model, the nanoplatform achieved a 100% survival rate for up to 60 days. Immunological analysis revealed each 2.36-fold, 4.19-fold increase in activated dendritic cells and Tc cells respectively, and significant reductions in MDSCs (0.48-fold) and regulatory T cells (0.5-fold). As a result, this study demonstrates the potential of the synergistic photoimmuno-lure nanoplatform as a clinical solution to overcome the immunosuppressive TME of GBM and activate innate and adaptive immunity for effective treatment.
Collapse
Affiliation(s)
- Minji Ahn
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- Department of Radiology, Feinburg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Yuhyun Na
- NBR incorporation, BI 205, 43 Jibong-ro, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Hani Choi
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Sanghee Lee
- Department of Radiology, Feinburg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jangsu Lee
- Department of Biomedical Science, Institute of Cell and Gene Therapy, CHA University, CHA Biocomplex, 335 Pangyo-ro, Sampyeong-Dong, Bundang-gu, Seongnam-si, Gyeonggi-Do, 13488, Republic of Korea
| | - Soon A Park
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| |
Collapse
|
4
|
Kawauchi D, Narita Y. The curse of blood-brain barrier and blood-tumor barrier in malignant brain tumor treatment. Int J Clin Oncol 2025:10.1007/s10147-025-02777-3. [PMID: 40338447 DOI: 10.1007/s10147-025-02777-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025]
Abstract
The blood-brain barrier (BBB) is crucial for brain homeostasis but is a major obstacle in delivering anticancer drugs to brain tumors. However, this perspective requires re-evaluation, particularly for malignant brain tumors, such as gliomas and brain metastases. In these aggressive tumors, the BBB undergoes significant alterations, leading to the formation of a more permeable blood-tumor barrier. While this increased permeability allows better drug penetration, heterogeneity in blood-tumor barrier (BTB) integrity across different tumor regions remains a challenge. Additionally, the main challenge in treating brain tumors lies not in BBB penetration but in the lack of effective drugs. Conventional chemotherapies, including temozolomide and nitrosourea agents, have shown limited efficacy, and resistance mechanisms often reduce their long-term benefits. The "BBB curse" has often been blamed for the slow progress in drug development. However, emerging evidence suggests that even larger-molecule therapies, such as antibody-drug conjugates, can successfully target brain tumors. This review aims to critically reassess the roles of the BBB and BTB in brain tumor therapy, highlighting their impact on drug delivery and evaluating the current landscape of chemotherapeutic strategies. Furthermore, it explores new approaches to overcome treatment limitations, emphasizing the need for personalized and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Daisuke Kawauchi
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo City, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo City, Japan.
| |
Collapse
|
5
|
DePalma T, Rodriguez M, Kollin L, Hughes K, Jones K, Stagner E, Venere M, Skardal A. A Microfluidic Blood Brain Barrier Model to Study the Influence of Glioblastoma Tumor Cells on BBB Function. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411361. [PMID: 40183747 DOI: 10.1002/smll.202411361] [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: 11/26/2024] [Revised: 02/13/2025] [Indexed: 04/05/2025]
Abstract
The blood brain barrier (BBB) plays an essential role in regulating brain function by controlling the transport of nutrients and preventing toxins from moving from the rest of the body's circulation into the brain. Because it is more selective than most other endothelial barriers, many therapeutic candidates fail to cross the BBB, making it difficult to design novel drugs to treat many pathologies in the brain. In addition, BBB dysfunction is observed in many brain diseases including glioblastoma (GB), an aggressive, universally fatal primary brain tumor. Here, a novel 3D microfluidic model of the BBB is designed using human cells and a brain-mimetic hydrogel. The in vitro BBB model replicates several key functions of the human BBB. This system has low permeability to small molecules and responds to inflammatory cues. The addition of GB cells to the model reveals that BBB function changes in a tumor-cell-population-dependent manner. Some GB cell populations lead to increased diffusive permeability while others induce increased immune cell binding. Together, these results indicate that this model can be used to investigate disease progression and drug delivery in GB.
Collapse
Affiliation(s)
- Thomas DePalma
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Marco Rodriguez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Luke Kollin
- Department of Radiation Oncology, Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Kennedy Hughes
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Katie Jones
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Emerie Stagner
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Monica Venere
- Department of Radiation Oncology, Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
6
|
Sarkaria JN, Kizilbash SH, Giannini C. Veliparib Plus Temozolomide for MGMT-Methylated Glioblastoma-Reply. JAMA Oncol 2025; 11:569-570. [PMID: 40079938 DOI: 10.1001/jamaoncol.2025.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
|
7
|
Shenoy G, Sahu A, Kuhn M, Khristov V, Heebner M, Wilding H, Clegg T, Bhanja D, Wade Q, Liermann LJ, Wang D, Smith N, Remite-Berthet G, Khunsriraksakul C, Palsa K, Slagle-Webb B, Mansouri A, Zacharia BE, Proctor EA, Connor JR. Analysis of transition metal content in glioblastoma reveals association between iron and survival. Transl Oncol 2025; 55:102376. [PMID: 40163909 PMCID: PMC11993186 DOI: 10.1016/j.tranon.2025.102376] [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: 11/28/2024] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
INTRODUCTION Little is known about the role of transition metals in glioblastoma progression. Here we investigated whether transition metal content is associated with glioblastoma outcomes. METHODS Tumor samples were obtained from 37 newly diagnosed patients with glioblastoma, 21 of which had matched plasma. Iron, zinc, manganese, and copper content in those samples was quantified via inductively-coupled mass spectrometry or atomic emission spectrometry, and subsequently analyzed for associations with overall survival. Multiplexed immune profiling was performed to determine whether transition metal content was associated with altered cytokine profiles. RESULTS Higher plasma iron levels were strongly associated with prolonged survival (Kaplan-Meier analysis: 30.15 months vs. 12.43 months, P = 0.0036; Multivariate Cox regression analysis: HR: 0.79 [0.64 - 0.97], P = 0.03). Zinc, manganese, and copper concentration in plasma or tumor and iron in tumor were not significantly associated with overall survival. Immune profiling of plasma and tumor samples revealed that plasma iron correlated with plasma IFN-β concentration (R = 0.63, P = 0.0057) in patients with glioblastoma. No correlation of plasma iron and IFN-β was observed in age- and sex- matched healthy individuals (R = -0.15, P = 0.153). Plasma transition metal concentration did not correlate with tumor transition metal concentration. Within tumors, manganese and zinc were correlated (R = 0.52, P = 0.0048) as well as copper and zinc (R = 0.36, P = 0.038). CONCLUSIONS Plasma iron is associated with survival in glioblastoma patients and may serve as a prognostic marker. The mechanisms underlying this association require further study.
Collapse
Affiliation(s)
- Ganesh Shenoy
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States
| | - Aurosman Sahu
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States
| | - Madison Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States
| | - Vladimir Khristov
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States
| | | | - Hannah Wilding
- Penn State College of Medicine, Hershey, PA, United States
| | - Taylor Clegg
- Penn State College of Medicine, Hershey, PA, United States
| | | | - Quinn Wade
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States
| | - Laura J Liermann
- Laboratory for Isotopes and Metals in the Environment, Pennsylvania State University, University Park, PA, United States
| | - Dongxiang Wang
- Laboratory for Isotopes and Metals in the Environment, Pennsylvania State University, University Park, PA, United States
| | - Nataliya Smith
- Penn State College of Medicine, Hershey, PA, United States
| | | | | | - Kondaiah Palsa
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States
| | - Becky Slagle-Webb
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States
| | - Brad E Zacharia
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States; Department of Pharmacology, Penn State College of Medicine, Hershey, PA, United States; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, United States; Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, United States; Penn State Neuroscience Institute, Penn State College of Medicine, Hershey, PA, United States; Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, United States
| | - James R Connor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, United States.
| |
Collapse
|
8
|
Bhutani B, Sharma V, Ganguly NK, Rana R. Unravelling the modified T cell receptor through Gen-Next CAR T cell therapy in Glioblastoma: Current status and future challenges. Biomed Pharmacother 2025; 186:117987. [PMID: 40117901 DOI: 10.1016/j.biopha.2025.117987] [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: 12/23/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025] Open
Abstract
PURPOSE Despite current technological advancements in the treatment of glioma, immediate alleviation of symptoms can be catered by therapeutic modalities, including surgery, chemotherapy, and combinatorial radiotherapy that exploit aberrations of glioma. Additionally, a small number of target antigens, their heterogeneity, and immune evasion are the potential reasons for developing targeted therapies. This oncologic milestone has catalyzed interest in developing immunotherapies against Glioblastoma to improve overall survival and cure patients with high-grade glioma. The next-gen CAR-T Cell therapy is one of the effective immunotherapeutic strategies in which autologous T cells have been modified to express receptors against GBM and it modulates cytotoxicity. METHODS In this review article, we examine preclinical and clinical outcomes, and limitations as well as present cutting-edge techniques to improve the function of CAR-T cell therapy and explore the possibility of combination therapy. FINDINGS To date, several CAR T-cell therapies are being evaluated in clinical trials for GBM and other brain malignancies and multiple preclinical studies have demonstrated encouraging outcomes. IMPLICATIONS CAR-T cell therapy represents a promising therapeutic paradigm in the treatment of solid tumors but a few limitations include, the blood-brain barrier (BBB), antigen escape, tumor microenvironment (TME), tumor heterogeneity, and its plasticity that suppresses immune responses weakens the ability of this therapy. Additional investigation is required that can accurately identify the targets and reflect the similar architecture of glioblastoma, thus optimizing the efficiency of CAR-T cell therapy; allowing for the selection of patients most likely to benefit from immuno-based treatments.
Collapse
Affiliation(s)
- Bhavya Bhutani
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Vyoma Sharma
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India.
| |
Collapse
|
9
|
Ali MF, Riviere-Cazaux C, Johnson SH, Salvatori R, Penheiter AR, Smadbeck JB, Murphy SJ, Harris FR, McCune LF, Carlstrom LP, Barrett MT, Kosari F, Jones LA, Ida C, Borad MJ, Bendok BR, Quiñones-Hinojosa A, Porter AB, Mrugala MM, Jaeckle KA, Anastasiadis PZ, Kizilbash SH, Cheville JC, Routman DM, Burns TC, Vasmatzis G. Personalized Tumor-Specific Amplified DNA Junctions in Peripheral Blood of Patients with High-Grade Gliomas. Clin Cancer Res 2025; 31:1700-1710. [PMID: 40019927 PMCID: PMC12010965 DOI: 10.1158/1078-0432.ccr-24-3233] [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: 09/27/2024] [Revised: 12/30/2024] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
PURPOSE Monitoring disease progression in patients with high-grade gliomas (HGG) is challenging due to treatment-related changes in imaging and the requirement for neurosurgical intervention to obtain diagnostic tissue. DNA junctions in HGG often amplify oncogenes, making these DNA fragments potentially more abundant in blood than monoallelic mutations. In this study, we piloted a cell-free DNA approach for disease detection in the plasma of patients with HGG by leveraging patient-specific DNA junctions associated with oncogene amplifications. EXPERIMENTAL DESIGN Whole-genome sequencing of grade 3 or 4 isocitrate dehydrogenase-mutant or wild-type astrocytomas was utilized to identify amplified junctions. Individualized qPCR assays were developed using patient-specific primers designed for the amplified junction. ctDNA levels containing these junctions were measured in patient plasma samples. RESULTS Unique amplified junctions were evaluated by individualized semi-qPCR assays in presurgical plasma of 18 patients, 15 with tumor-associated focal amplifications and three without tumor-associated focal amplifications. high copy-number junctions were robustly detected in the plasma of 14 of 15 (93.3%) patients with amplified junctions and none of the controls. Changes in junction abundance correlated with disease trajectory in serial plasma samples from five patients, including increased abundance of amplified junctions preceding radiographic disease progression. CONCLUSIONS In patients with grade 3 or 4 astrocytomas who had tumor-associated amplifications, patient-specific amplified junctions were successfully detected in assayed plasma from most patients. Longitudinal analysis of plasma samples correlated with disease trajectory, including cytoreduction and progression.
Collapse
Affiliation(s)
- Mohamed F. Ali
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Sarah H. Johnson
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Rebecca Salvatori
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Lab Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Alan R. Penheiter
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - James B. Smadbeck
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Stephen J. Murphy
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Faye R. Harris
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Lex F. McCune
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Michael T. Barrett
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona
| | - Farhad Kosari
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Leila A. Jones
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Cristiane Ida
- Department of Lab Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Mitesh J. Borad
- Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Bernard R. Bendok
- Department of Neurological Surgery, Mayo Clinic, Phoenix, Arizona
- Neurosurgery Simulation and Innovation Laboratory, Mayo Clinic, Phoenix, Arizona
- Precision Neuro-Therapeutics Innovation Laboratory, Mayo Clinic, Phoenix, Arizona
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
- Department of Otolaryngology–Head and Neck Surgery, Mayo Clinic, Phoenix, Arizona
| | | | | | | | | | | | | | - John C. Cheville
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Lab Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - David M. Routman
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Terry C. Burns
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota
| | - George Vasmatzis
- Center for Individualized Medicine and Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
10
|
Khot S, Mahajan U, Jadhav A, Vaishampayan P, Bagul U, Gadhave D, Gorain B, Kokare C. Nose-to-brain delivery of sorafenib-loaded lipid-based poloxamer-carrageenan nanoemulgel: Formulation and therapeutic investigation in glioblastoma-induced orthotopic rat model. Int J Biol Macromol 2025; 309:142861. [PMID: 40188927 DOI: 10.1016/j.ijbiomac.2025.142861] [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: 12/04/2024] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Glioblastoma multiforme (GBM) has a poor clinical prognosis, where conventional treatment offers therapeutic limitations. Therefore, the current study introduces a first-of-its-kind sorafenib (SOR) nanoemulsion (SNE) loaded with poloxamer-carrageenan nanoemulgel (SPCNEG), a novel dual-functional and natural polymer-based payload system for effective intranasal chemotherapeutic administration. The nanoformulation was developed using carrageenan (a natural gelling agent), poloxamer (a mucoadhesive agent), glyceryl caprate as lipid, and Cremophor EL:PEG 400 blend as surfactant system. The improved biopharmaceutical attributes of developed formulations were confirmed from the release experiments, revealing augmentation in drug release from SNE (84.56 ± 3.78 %) and SPCNEG (68.62 ± 4.11 %) up to 3.41- and 8.12-fold compared to plain SOR. The ex vivo experiments showed a similar enhancement in drug permeation. Moreover, the SNE also showed superior performance on glioma cell lines, as indicated by lower IC50 (2.23 μg/mL) than plain SOR (16.61 μg/mL). The pharmacokinetic study revealed a 2.52- and 3.24-fold increase in SNE and SPCNEG brain concentration, respectively, compared to Soranib®. Additionally, a high correlation was also observed between in vitro drug release and in vivo absorption at prespecified time intervals for developed formulations. In conclusion, the current research promising and non-invasive alternative to existing interventions for enhanced brain targeting potential.
Collapse
Affiliation(s)
- Shubham Khot
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| | - Unmesh Mahajan
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| | - Amol Jadhav
- Institute of Applied Biological Research and Development, (IABRD), A division of Nirav Biosolutions Pvt Ltd, Aundh, Pune 411007, Maharashtra, India
| | | | - Uddhav Bagul
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| | - Dnyandev Gadhave
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Chandrakant Kokare
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India.
| |
Collapse
|
11
|
Rastegar G, Salman MM, Sirsi SR. Oxygen Sensitive Drug Release Using Hemoglobin Microbubbles: A New Approach to Targeting Hypoxia in Ultrasound-Mediated Drug Delivery. Int J Pharm 2025; 675:125521. [PMID: 40139453 DOI: 10.1016/j.ijpharm.2025.125521] [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: 01/03/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Targeted drug delivery strategies using focused ultrasound (FUS) are gaining prominence in clinical application. FUS offers deep tissue penetration and precise targeting capabilities. The capabilities of FUS in targeted drug delivery are greatly enhanced by the introduction of ultrasound contrast agents (UCAs - also known as microbubbles). This study introduces a novel hypoxia-targeting drug delivery system using hemoglobin microbubbles (HbMBs) conjugated with doxorubicin-loaded liposomes (LDOX). Previously, we reported that HbMBs exhibit significant acoustic response differences between oxygenated and deoxygenated environments due to hemoglobin's conformational changes, which alters the MBs' shell elasticity as well as resonance frequency. In this study, we coated the surface of HBMBs with LDOX to create Lip-HBMB complex and subsequently investigated its drug release at different oxygen partial pressures (pO2) when exposed to an ultrasound field. Results showed significantly higher drug release at lower oxygen levels, with about 10-times higher release at 5 mmHg pO2 than 160 mmHg pO2 at 0.5 W/cm2 US intensity and 3 MHz frequency. This highlights Lip-HbMBs' potential for targeted drug delivery to hypoxic tumor regions, marking a significant advancement in focused ultrasound-mediated drug delivery. This study marks the first-ever report of ultrasound-mediated oxygen-sensitive drug uncaging, which holds promise in enhancing FUS specificity and addressing the challenges posed by metastatic cancer.
Collapse
Affiliation(s)
- Ghazal Rastegar
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Mohammad Musa Salman
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA.
| |
Collapse
|
12
|
Afshar Y, Sharifi N, Kamroo A, Yazdanpanah N, Saleki K, Rezaei N. Implications of glioblastoma-derived exosomes in modifying the immune system: state-of-the-art and challenges. Rev Neurosci 2025; 36:315-325. [PMID: 39528347 DOI: 10.1515/revneuro-2024-0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma is a brain cancer with a poor prognosis. Failure of classical chemotherapy and surgical treatments indicates that new therapeutic approaches are needed. Among cell-free options, exosomes are versatile extracellular vesicles (EVs) that carry important cargo across barriers such as the blood-brain barrier (BBB) to their target cells. This makes exosomes an interesting option for the treatment of glioblastoma. Moreover, exosomes can comprise many therapeutic cargos, including lipids, proteins, and nucleic acids, sampled from special intercellular compartments of their origin cell. Cells exposed to various immunomodulatory stimuli can generate exosomes enriched in specific therapeutic molecules. Notably, the secretion of exosomes could modify the immune response in innate and adaptive immune systems. For instance, glioblastoma-associated exosomes (GBex) uptake by macrophages could influence macrophage dynamics (e.g., shifting CD markers expression). Expression of critical immunoregulatory proteins such as cytotoxic T-lymphocyte antigen-1 (CTLA1) and programmed death-1 (PD-1) on GBex indicates the direct crosstalk of these nano-size vesicles with the immune system. The present study reviews the role of exosomes in immune system cells, including B cells, T cells, natural killer (NK) cells, and dendritic cells (DCs), as well as novel technologies in the field.
Collapse
Affiliation(s)
- Yashmin Afshar
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Negin Sharifi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- Faculty of Medicine, Tehran Medical Science Branch, Islamic Azad University, Tehran, 1584743311, Iran
| | - Amirhossein Kamroo
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Students' Scientific Research Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
| | - Niloufar Yazdanpanah
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
| | - Kiarash Saleki
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- Student Research Committee, Babol University of Medical Sciences, Babol, 4717647745, Iran
- USERN Office, Babol University of Medical Sciences, Babol, 4717647745, Iran
- Department of E-Learning in Medical Sciences, Faculty of Medical Education and Learning Technologies, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, 1416634793, Iran
- Department of Immunology, School of Medicine, 48439 Tehran University of Medical Sciences , Tehran, 1416634793, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Stockholms, 10316, Sweden
| |
Collapse
|
13
|
Fuse H, Zheng Y, Alzoubi I, Graeber MB. TAMing Gliomas: Unraveling the Roles of Iba1 and CD163 in Glioblastoma. Cancers (Basel) 2025; 17:1457. [PMID: 40361384 PMCID: PMC12070867 DOI: 10.3390/cancers17091457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Gliomas, the most common type of primary brain tumor, are a significant cause of morbidity and mortality worldwide. Glioblastoma, a highly malignant subtype, is particularly common, aggressive, and resistant to treatment. The tumor microenvironment (TME) of gliomas, especially glioblastomas, is characterized by a distinct presence of tumor-associated macrophages (TAMs), which densely infiltrate glioblastomas, a hallmark of these tumors. This macrophage population comprises both tissue-resident microglia as well as macrophages derived from the walls of blood vessels and the blood stream. Ionized calcium-binding adapter molecule 1 (Iba1) and CD163 are established cellular markers that enable the identification and functional characterization of these cells within the TME. This review provides an in-depth examination of the roles of Iba1 and CD163 in malignant gliomas, with a focus on TAM activation, migration, and immunomodulatory functions. Additionally, we will discuss how recent advances in AI-enhanced cell identification and visualization techniques have begun to transform the analysis of TAMs, promising unprecedented precision in their characterization and providing new insights into their roles within the TME. Iba1 and CD163 appear to have both unique and shared roles in glioma pathobiology, and both have the potential to be targeted through different molecular and cellular mechanisms. We discuss the therapeutic potential of Iba1 and CD163 based on available preclinical (experimental) and clinical (human tissue-based) evidence.
Collapse
Affiliation(s)
- Haneya Fuse
- School of Medicine, Sydney Campus, University of Notre Dame, 160 Oxford Street, Sydney, NSW 2010, Australia;
| | - Yuqi Zheng
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia;
| | - Islam Alzoubi
- School of Computer Science, The University of Sydney, J12/1 Cleveland St, Sydney, NSW 2008, Australia;
| | - Manuel B. Graeber
- Ken Parker Brain Tumor Research Laboratories, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2050, Australia;
- University of Sydney Association of Professors (USAP), University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
14
|
Janowicz PW, Boele T, Maschmeyer RT, Gholami YH, Kempe EG, Stringer BW, Stoner SP, Zhang M, du Toit-Thompson T, Williams F, Touffu A, Munoz L, Kuncic Z, Brighi C, Waddington DEJ. Enhanced detection of glioblastoma vasculature with superparamagnetic iron oxide nanoparticles and MRI. Sci Rep 2025; 15:14283. [PMID: 40274951 PMCID: PMC12022243 DOI: 10.1038/s41598-025-97943-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Detecting glioblastoma infiltration in the brain is challenging due to limited MRI contrast beyond the enhancing tumour core. This study aims to investigate the potential of superparamagnetic iron oxide nanoparticles (SPIONs) as contrast agents for improved detection of diffuse brain cancer. We examine the distribution and pharmacokinetics of SPIONs in glioblastoma models with intact and disrupted blood-brain barriers. Using MRI, we imaged RN1-luc and U87MG mice injected with Gadovist and SPIONs, observing differences in blood-brain barrier permeability. Peripheral imaging showed strong uptake of nanoparticles in the liver and spleen, while vascular and renal signals were transient. Susceptibility gradient mapping enabled positive nanoparticle contrast within tumours and provided additional information on tumour angiogenesis. This approach offers a novel method for detecting diffuse brain cancer. Our findings demonstrate that SPIONs enhance glioblastoma detection beyond conventional MRI, providing insights into tumour angiogenesis and opening new avenues for early diagnosis and targeted treatment strategies.
Collapse
Affiliation(s)
- Phillip W Janowicz
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| | - Thomas Boele
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Richard T Maschmeyer
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
| | - Yaser H Gholami
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, Australia
| | - Emma G Kempe
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Brett W Stringer
- Institute for Biomedicine and Glycomics, Griffith University, Brisbane, Australia
| | - Shihani P Stoner
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Marie Zhang
- Imagion Biosystems Ltd, Melbourne, Australia
| | - Taymin du Toit-Thompson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Fern Williams
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Aude Touffu
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Lenka Munoz
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Zdenka Kuncic
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
- The Sydney Nano Institute, The University of Sydney, Sydney, Australia
| | - Caterina Brighi
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - David E J Waddington
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
15
|
Roy AA, Pandey A, Dhas N, Hegde MM, Parekh HS, Andugulapati SB, Nandakumar K, Satish Rao BS, Mutalik S. The Confluence of Nanotechnology and Heat Shock Protein 70 in Pioneering Glioblastoma Multiforme Therapy: Forging Pathways Towards Precision Targeting and Transformation. Adv Pharmacol Pharm Sci 2025; 2025:1847197. [PMID: 40313865 PMCID: PMC12045689 DOI: 10.1155/adpp/1847197] [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: 08/13/2024] [Accepted: 03/01/2025] [Indexed: 05/03/2025] Open
Abstract
Heat-shock protein 70 (HSP70) and nanotechnology have emerged as promising avenues in glioblastoma multiforme (GBM) therapy, addressing the critical challenges posed by its aggressive nature and therapeutic resistance. HSP70's dual role in cellular stress response and tumour survival emphasises its potential as both a biomarker and therapeutic target. This review explores the innovative integration of HSP70 with nanotechnology, emphasising advancements in imaging, drug delivery and combination therapies. Nanoparticles, including SPIONs, liposomes, gold nanoparticles and metal-organic frameworks, demonstrate enhanced targeting and therapeutic efficacy through HSP70 modulation. Functionalized nanocarriers exploit HSP70's tumour-specific overexpression to improve drug delivery, minimise off-target effects and overcome the blood-brain barrier. Emerging strategies such as chemophototherapy, immunotherapy and photothermal therapy leverage HSP70's interactions within the tumour microenvironment, enabling synergistic treatment modalities. The review also highlights translational challenges, including heterogeneity of GBM, regulatory hurdles and variability in the enhanced permeability and retention (EPR) effect. Integrating computational modelling, personalised approaches and adaptive trial designs is crucial for clinical translation. By bridging nanotechnology and molecular biology, HSP70-targeted strategies hold transformative potential to redefine GBM diagnosis and treatment, offering hope for improved survival and quality of life. Trial Registration: ClinicalTrials.gov identifier: NCT00054041 and NCT04628806.
Collapse
Affiliation(s)
- Amrita Arup Roy
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Abhijeet Pandey
- Global Drug Development/Technical Research and Development, Novartis Healthcare Pvt. Ltd., Genome Valley, Hyderabad 500081, Telangana, India
| | - Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Manasa Manjunath Hegde
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Harendra S. Parekh
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Sai Balaji Andugulapati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, Telangana, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Bola Sadashiva Satish Rao
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
16
|
Salbini M, Formato A, Mongiardi MP, Levi A, Falchetti ML. Kinase-Targeted Therapies for Glioblastoma. Int J Mol Sci 2025; 26:3737. [PMID: 40332381 PMCID: PMC12027600 DOI: 10.3390/ijms26083737] [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: 03/04/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Protein phosphorylation and dephosphorylation are key mechanisms that regulate cellular activities. The addition or removal of phosphate groups by specific enzymes, known as kinases and phosphatases, activates or inhibits many enzymes and receptors involved in various cell signaling pathways. Dysregulated activity of these enzymes is associated with various diseases, predominantly cancers. Synthetic and natural single- and multiple-kinase inhibitors are currently being used as targeted therapies for different tumors, including glioblastoma. Glioblastoma IDH-wild-type is the most aggressive brain tumor in adults, with a median overall survival of 15 months. The great majority of glioblastoma patients present mutations in receptor tyrosine kinase (RTK) signaling pathways responsible for tumor initiation and/or progression. Despite this, the multi-kinase inhibitor regorafenib has only recently been approved for glioblastoma patients in some countries. In this review, we analyze the history of kinase inhibitor drugs in glioblastoma therapy.
Collapse
Affiliation(s)
| | | | | | | | - Maria Laura Falchetti
- Institute of Biochemistry and Cell Biology, National Research Council, Via Ercole Ramarini 32, Monterotondo, 00015 Rome, Italy; (M.S.); (A.F.); (M.P.M.); (A.L.)
| |
Collapse
|
17
|
Shamul JG, Wang Z, Gong H, Ou W, White AM, Moniz-Garcia DP, Gu S, Clyne AM, Quiñones-Hinojosa A, He X. Meta-analysis of the make-up and properties of in vitro models of the healthy and diseased blood-brain barrier. Nat Biomed Eng 2025; 9:566-598. [PMID: 39304761 PMCID: PMC11922799 DOI: 10.1038/s41551-024-01250-2] [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: 07/06/2023] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
In vitro models of the human blood-brain barrier (BBB) are increasingly used to develop therapeutics that can cross the BBB for treating diseases of the central nervous system. Here we report a meta-analysis of the make-up and properties of transwell and microfluidic models of the healthy BBB and of BBBs in glioblastoma, Alzheimer's disease, Parkinson's disease and inflammatory diseases. We found that the type of model, the culture method (static or dynamic), the cell types and cell ratios, and the biomaterials employed as extracellular matrix are all crucial to recapitulate the low permeability and high expression of tight-junction proteins of the BBB, and to obtain high trans-endothelial electrical resistance. Specifically, for models of the healthy BBB, the inclusion of endothelial cells and pericytes as well as physiological shear stresses (~10-20 dyne cm-2) are necessary, and when astrocytes are added, astrocytes or pericytes should outnumber endothelial cells. We expect this meta-analysis to facilitate the design of increasingly physiological models of the BBB.
Collapse
Affiliation(s)
- James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Zhiyuan Wang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Hyeyeon Gong
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Alisa M White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | | | - Shuo Gu
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA
| | | | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA.
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
18
|
Roncali L, Hindré F, Samarut E, Lacoeuille F, Rousseau A, Lemée JM, Garcion E, Chérel M. Current landscape and future directions of targeted-alpha-therapy for glioblastoma treatment. Theranostics 2025; 15:4861-4889. [PMID: 40303349 PMCID: PMC12036880 DOI: 10.7150/thno.106081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/02/2025] [Indexed: 05/02/2025] Open
Abstract
Glioblastoma (GB) is the most aggressive malignancy of the central nervous system. Despite two decades of intensive research since the establishment of the standard of care, emerging strategies have yet to produce consistent satisfactory outcomes. Because of its specific localisation and intricate characteristics, GB is a uniquely regulated solid tumour with a strong resistance to therapy. Advances in targeted radionuclide therapy (TRT), particularly with the introduction of a-emitting radionuclides, have unveiled potential avenues for the management of GB. Recent preclinical and clinical studies underscored promising advancements for targeted-α-therapy (TAT), but these therapeutic approaches exhibit a vast design heterogeneity, encompassing diverse radionuclides, vectors, target molecules, and administration modalities. This review seeks to critically assess the therapeutic landscape of GB through the perspective of TAT. Here, the focus is made on the advancements and limitations of in vivo explorations, pilot studies, and clinical trials, to determine the best directions for future investigations. In doing so, we hope to identify existing challenges and draw insights that might pave the way towards a more effective therapeutic approach.
Collapse
Affiliation(s)
- Loris Roncali
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela; E-15782 Santiago de Compostela, Spain
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
| | - François Hindré
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- PRIMEX (Experimental Imagery and Radiobiology Platform), University of Angers, SFR 4208; F-49000 Angers, France
| | - Edouard Samarut
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
- Department of Neurosurgery & Neurotraumatology, University Hospital of Nantes; F-44093 Nantes, France
| | - Franck Lacoeuille
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Nuclear Medicine, University Hospital of Angers; F-49000 Angers, France
| | - Audrey Rousseau
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Pathology, University Hospital of Angers; F-49000 Angers, France
| | - Jean-Michel Lemée
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Neurosurgery, University Hospital of Angers; F-49000 Angers, France
| | - Emmanuel Garcion
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- PACEM (Platform of Cellular and Molecular Analysis), University of Angers, SFR 4208; F-49000 Angers, France
| | - Michel Chérel
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
- Institut de Cancérologie de l'Ouest, Department of Nuclear Medicine; F-44160 Saint-Herblain, France
| |
Collapse
|
19
|
Ożarowski M, Karpiński TM, Czerny B, Kamiński A, Seremak-Mrozikiewicz A. Plant Alkaloids as Promising Anticancer Compounds with Blood-Brain Barrier Penetration in the Treatment of Glioblastoma: In Vitro and In Vivo Models. Molecules 2025; 30:1561. [PMID: 40286187 PMCID: PMC11990316 DOI: 10.3390/molecules30071561] [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: 12/30/2024] [Revised: 03/08/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
Glioblastoma (GBM) is one of the most invasive central nervous system tumors, with rising global incidence. Therapy resistance and poor prognosis highlight the urgent need for new anticancer drugs. Plant alkaloids, a largely unexplored yet promising class of compounds, have previously contributed to oncology treatments. While past reviews provided selective insights, this review aims to collectively compare data from the last decade on (1) plant alkaloid-based anticancer drugs, (2) alkaloid transport across the blood-brain barrier (BBB) in vitro and in vivo, (3) alkaloid mechanisms of action in glioblastoma models (in vitro, in vivo, ex vivo, and in silico), and (4) cytotoxicity and safety profiles. Additionally, innovative drug delivery systems (e.g., nanoparticles and liposomes) are discussed. Focusing on preclinical studies of single plant alkaloids, this review includes 22 botanical families and 28 alkaloids that demonstrated anti-GBM activity. Most alkaloids act in a concentration-dependent manner by (1) reducing glioma cell viability, (2) suppressing proliferation, (3) inhibiting migration and invasion, (4) inducing cell death, (5) downregulating Bcl-2 and key signaling pathways, (6) exhibiting antiangiogenic effects, (7) reducing tumor weight, and (8) improving survival rates. The toxic and adverse effect analysis suggests that alkaloids such as noscapine, lycorine, capsaicin, chelerythrine, caffeine, boldine, and colchicine show favorable therapeutic potential. However, tetrandrine, nitidine, harmine, harmaline, cyclopamine, cocaine, and brucine may pose greater risks than benefits. Piperine's toxicity and berberine's poor bioavailability suggest the need for novel drug formulations. Several alkaloids (kukoamine A, cyclovirobuxine D, α-solanine, oxymatrine, rutaecarpine, and evodiamine) require further pharmacological and toxicological evaluation. Overall, while plant alkaloids show promise in glioblastoma therapy, progress in assessing their BBB penetration remains limited. More comprehensive studies integrating glioma research and advanced drug delivery technologies are needed.
Collapse
Affiliation(s)
- Marcin Ożarowski
- Department of Biotechnology, Institute of Natural Fibres and Medicinal Plants—National Research Institute, Wojska Polskiego 71b, 60-630 Poznań, Poland
| | - Tomasz M. Karpiński
- Chair and Department of Medical Microbiology, Poznań University of Medical Sciences, Rokietnicka 10, 60-806 Poznań, Poland
| | - Bogusław Czerny
- Department of General Pharmacology and Pharmacoeconomics, Pomeranian Medical University in Szczecin, Żołnierska 48, 70-204 Szczecin, Poland;
- Institute of Natural Fibres and Medicinal Plants—National Research Institute, Wojska Polskiego 71b, 60-630 Poznań, Poland
| | - Adam Kamiński
- Department of Orthopaedics and Traumatology, Independent Public Clinical Hospital No. 1, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Agnieszka Seremak-Mrozikiewicz
- Division of Perinatology and Women’s Disease, Poznań University of Medical Sciences, Polna 33, 60-535 Poznań, Poland;
- Laboratory of Molecular Biology in Division of Perinatology and Women’s Diseases, University of Medical Sciences, Polna 33, 60-535 Poznań, Poland
| |
Collapse
|
20
|
Keshari R, Dewani M, Kaur N, Patel GK, Singh SK, Chandra P, Prasad R, Srivastava R. Lipid Nanocarriers as Precision Delivery Systems for Brain Tumors. Bioconjug Chem 2025; 36:347-366. [PMID: 39937652 DOI: 10.1021/acs.bioconjchem.5c00007] [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: 02/14/2025]
Abstract
Brain tumors, particularly glioblastomas, represent the most complicated cancers to treat and manage due to their highly invasive nature and the protective barriers of the brain, including the blood-brain barrier (BBB). The efficacy of currently available treatments, viz., radiotherapy, chemotherapy, and immunotherapy, are frequently limited by major side effects, drug resistance, and restricted drug penetration into the brain. Lipid nanoparticles (LNPs) have emerged as a promising and targeted delivery system for brain tumors. Lipid nanocarriers have gained tremendous attention for brain tumor therapeutics due to multiple drug encapsulation abilities, controlled release, better biocompatibility, and ability to cross the BBB. Herein, a detailed analysis of the design, mechanisms, and therapeutic benefits of LNPs in brain tumor treatment is discussed. Moreover, we also discuss the safety issues and clinical developments of LNPs and their current and future challenges. Further, we also focused on the clinical transformation of LNPs in brain tumor therapy by eliminating side effects and engineering the LNPs to overcome the related biological barriers, which provide personalized, affordable, and low-risk treatment options.
Collapse
Affiliation(s)
- Roshan Keshari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Mahima Dewani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Navneet Kaur
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - Girijesh Kumar Patel
- Cancer and Stem Cell Laboratory, Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India-211004
| | - Sumit Kumar Singh
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh 221005, India
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh 221005, India
| | - Rajendra Prasad
- School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh 221005, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
21
|
Zhang X, Artz N, Steindler DA, Hingtgen S, Satterlee AB. Exosomes: Traversing the blood-brain barrier and their therapeutic potential in brain cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189300. [PMID: 40097050 DOI: 10.1016/j.bbcan.2025.189300] [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: 10/01/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
The blood-brain barrier (BBB) presents a major challenge for the effective delivery of therapeutic agents to the brain tumor cells from the peripheral blood circulation, making the treatment of central nervous system (CNS)-related cancers more difficult and resistant to both standard treatments and emerging therapies. Exosomes, which serve as messengers for intercellular communication throughout the body, can naturally or be modified to penetrate the BBB. Recently, exosomes have been increasingly explored as an invasive or non-invasive approach for delivering therapeutic agents to the CNS. With their low immunogenicity, ease of modification, excellent cargo protection, and inherent ability to cross the BBB, exosomes hold great promise for revolutionizing targeted therapy for CNS-related diseases, including brain cancer. In this review, we highlight recent discoveries and insights into the mechanisms exosomes use to penetrate the BBB, the methods they employ to payload diverse therapeutics, and their roles in transporting therapeutic compounds for brain cancer and other neurological disorders.
Collapse
Affiliation(s)
- Xiaopei Zhang
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Nichole Artz
- Department of Pediatric Hematology/Oncology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Dennis A Steindler
- Steindler Consulting, Boston, MA, USA; Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Shawn Hingtgen
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew Benson Satterlee
- Eshelman School of Pharmacy, Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Eshelman Institute for Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
22
|
Rastegar-Pouyani N, Farzin MA, Zafari J, Haji Abdolvahab M, Hassani S. Repurposing the anti-parasitic agent pentamidine for cancer therapy; a novel approach with promising anti-tumor properties. J Transl Med 2025; 23:258. [PMID: 40033361 PMCID: PMC11877826 DOI: 10.1186/s12967-025-06293-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/23/2025] [Indexed: 03/05/2025] Open
Abstract
Pentamidine (PTM) is an aromatic diamidine administered for infectious diseases, e.g. sleeping sickness, malaria, and Pneumocystis jirovecii pneumonia. Due to similarities of cellular mechanisms between human cells and such infections, PTM has also been proposed for repurposing in non-infectious diseases such as cancer. Indeed, by modulating different signaling pathways such as PI3K/AKT, MAPK/ERK, p53, PD-1/PD-L1, etc., PTM has been shown to inhibit different properties of cancer, including proliferation, invasion, migration, hypoxia, and angiogenesis, while inducing anti-tumor immune responses and apoptosis. Given the promising implications of PTM for cancer treatment, however, the clinical translation of PTM in cancer is not without certain challenges. In fact, clinical trials have shown that systemic administration of PTM can be concurrent with serious adverse effects, e.g. hypoglycemia. Therefore, to reduce the administered doses of PTM, lower the risk of adverse effects, and prevent any potential drug resistance, while maintaining the anti-tumor efficacy, two main strategies have been suggested. One is combination therapy that employs PTM in conjunction with other anti-cancer modalities, such as chemotherapy and radiotherapy, and attacks tumor cells with significant additive or synergistic anti-tumor effects. The other is developing PTM-loaded nanocarrier drug delivery systems e.g. pegylated liposomes, chitosan-coated niosomes, squalene-based nanoparticles, hyaluronated lipid-polymer hybrid nanoparticles, etc., that offer enhanced pharmacokinetic characteristics, including increased bioavailability, sit-targeting, and controlled/sustained drug release. This review highlights the anti-tumor properties of PTM that favor its repurposing for cancer treatment, as well as, PTM-based combination therapies and nanocarrier delivery systems which can enhance therapeutic efficacy and simultaneously reduce toxicity.
Collapse
Affiliation(s)
- Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Tehran University of Medical Sciences, Tehran, Iran.
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Mohammad Amin Farzin
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Jaber Zafari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohadeseh Haji Abdolvahab
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Shokoufeh Hassani
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Li J, Wickramasinghe C, Jiang J, Wu A, Jiang Y, Tovmasyan A, Kim S, Sanai N. Mechanistic Modeling of Spatial Heterogeneity of Drug Penetration and Exposure in the Human Central Nervous System and Brain Tumors. Clin Pharmacol Ther 2025; 117:690-703. [PMID: 39575553 PMCID: PMC11835533 DOI: 10.1002/cpt.3505] [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: 08/29/2024] [Accepted: 11/08/2024] [Indexed: 02/20/2025]
Abstract
Direct measurement of spatial-temporal drug penetration and exposure in the human central nervous system (CNS) and brain tumors is difficult or infeasible. This study aimed to develop an innovative mechanistic modeling platform for quantitative prediction of spatial pharmacokinetics of systemically administered drugs in the human CNS and brain tumors. A nine-compartment CNS (9-CNS) physiologically-based pharmacokinetic model was developed to account for general anatomical structure and pathophysiological heterogeneity of the human CNS and brain tumors. Drug distribution into and within the CNS and tumors is driven by plasma concentration-time profiles and governed by drug properties and CNS pathophysiology. The model was validated by comparisons of model predictions and clinically observed data of six drugs (abemaciclib, ribociclib, pamiparib, olaparib, temuterkib, and ceritinib) in glioblastoma patients. As rigorously validated, the 9-CNS model allows reliable prediction of spatial pharmacokinetics in different regions of the brain parenchyma (i.e., parenchyma adjacent to CSF and deep parenchyma), tumors (i.e., tumor rim, bulk tumor, and tumor core), and CSF (i.e., ventricular CSF, cranial and spinal subarachnoid CSF). By considering inter-individual plasma pharmacokinetic variability and CNS/tumor heterogeneity, the model well predicts the inter-individual variability and spatial heterogeneity of drug exposure in the CNS and tumors as observed for all six drugs in glioblastoma patients. The 9-CNS model is a first-of-its kind, mechanism-based computational modeling platform that enables early reliable prediction of spatial CNS and tumor pharmacokinetics based on plasma concentration-time profiles. It provides a valuable tool to assist rational drug development and treatment for brain cancer.
Collapse
Affiliation(s)
- Jing Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | | | - Jun Jiang
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Andrew Wu
- Northville High School, 45700 Six Mile Rd, Northville, MI 48168
| | - Yuanyuan Jiang
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Artak Tovmasyan
- Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, AZ 85013
| | - Seongho Kim
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201
| | - Nader Sanai
- Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, AZ 85013
| |
Collapse
|
24
|
Wang L, Gu M, Zhang X, Kong T, Liao J, Zhang D, Li J. Recent Advances in Nanoenzymes Based Therapies for Glioblastoma: Overcoming Barriers and Enhancing Targeted Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413367. [PMID: 39854126 PMCID: PMC11905078 DOI: 10.1002/advs.202413367] [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: 10/21/2024] [Revised: 12/14/2024] [Indexed: 01/26/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor originating from glial cells, characterized by high recurrence rates and poor patient prognosis. The heterogeneity and complex biology of GBM, coupled with the protective nature of the blood-brain barrier (BBB), significantly limit the efficacy of traditional therapies. The rapid development of nanoenzyme technology presents a promising therapeutic paradigm for the rational and targeted treatment of GBM. In this review, the underlying mechanisms of GBM pathogenesis are comprehensively discussed, emphasizing the impact of the BBB on treatment strategies. Recent advances in nanoenzyme-based approaches for GBM therapy are explored, highlighting how these nanoenzymes enhance various treatment modalities through their multifunctional capabilities and potential for precise drug delivery. Finally, the challenges and therapeutic prospects of translating nanoenzymes from laboratory research to clinical application, including issues of stability, targeting efficiency, safety, and regulatory hurdles are critically analyzed. By providing a thorough understanding of both the opportunities and obstacles associated with nanoenzyme-based therapies, future research directions are aimed to be informed and contribute to the development of more effective treatments for GBM.
Collapse
Affiliation(s)
- Liyin Wang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Min Gu
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Xiaoli Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | | | - Jun Liao
- Institute of Systems BiomedicineBeijing Key Laboratory of Tumor Systems BiologySchool of Basic Medical SciencesPeking UniversityBeijing100191China
| | - Dan Zhang
- Shengjing Hospital of China Medical UniversityLiaoning110004China
| | - Jingwu Li
- The First Hospital of China Medical UniversityLiaoning110001China
| |
Collapse
|
25
|
Yang M, Wang X, Peng M, Wang F, Hou S, Xing R, Chen A. Nanomaterials Enhanced Sonodynamic Therapy for Multiple Tumor Treatment. NANO-MICRO LETTERS 2025; 17:157. [PMID: 39992547 PMCID: PMC11850698 DOI: 10.1007/s40820-025-01666-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/08/2025] [Indexed: 02/25/2025]
Abstract
Sonodynamic therapy (SDT) as an emerging modality for malignant tumors mainly involves in sonosensitizers and low-intensity ultrasound (US), which can safely penetrate the tissue without significant attenuation. SDT not only has the advantages including high precision, non-invasiveness, and minimal side effects, but also overcomes the limitation of low penetration of light to deep tumors. The cytotoxic reactive oxygen species can be produced by the utilization of sonosensitizers combined with US and kill tumor cells. However, the underlying mechanism of SDT has not been elucidated, and its unsatisfactory efficiency retards its further clinical application. Herein, we shed light on the main mechanisms of SDT and the types of sonosensitizers, including organic sonosensitizers and inorganic sonosensitizers. Due to the development of nanotechnology, many novel nanoplatforms are utilized in this arisen field to solve the barriers of sonosensitizers and enable continuous innovation. This review also highlights the potential advantages of nanosonosensitizers and focus on the enhanced efficiency of SDT based on nanosonosensitizers with monotherapy or synergistic therapy for deep tumors that are difficult to reach by traditional treatment, especially orthotopic cancers.
Collapse
Affiliation(s)
- Mengyao Yang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Xin Wang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Mengke Peng
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Fei Wang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Senlin Hou
- The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People's Republic of China.
| | - Ruirui Xing
- State Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China.
| | - Aibing Chen
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
26
|
Choi HJ, Han M, Jung B, Huh H, Lee EH, Choi JR, Park J. Evaluation of blood-tumor barrier permeability and doxorubicin delivery in rat brain tumor models using additional focused ultrasound stimulation. Sci Rep 2025; 15:6592. [PMID: 39994241 PMCID: PMC11850853 DOI: 10.1038/s41598-025-88379-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Focused ultrasound (FUS) has emerged as a promising technique for temporarily disrupting the blood-brain barrier (BBB) and blood-tumor barrier (BTB) to enhance the delivery of therapeutic agents. Despite its potential, optimizing FUS to maximize drug delivery while minimizing adverse effects remains a significant challenge. In this study, we evaluated a novel FUS protocol that incorporates additional FUS stimulation without microbubbles (MBs) ("FUS protocol") prior to conventional BBB disruption with MBs ("BBBD protocol") in a rat brain tumor model (n = 35). This approach aimed to validate its effectiveness in enhancing BBB/BTB disruption and facilitating doxorubicin delivery. T1-weighted contrast-enhanced and dynamic contrast-enhanced (DCE) MRI demonstrated significant increases in signal intensity and permeability (Ktrans) in the tumor region under the "FUS + BBBD protocol", with 2.65-fold and 2.08-fold increases, respectively, compared to the non-sonicated contralateral region. These values were also elevated compared to the conventional "BBBD protocol" by 1.45-fold and 1.25-fold, respectively. Furthermore, doxorubicin delivery in the targeted region increased by 1.91-fold under the "FUS + BBBD protocol", compared to a 1.44-fold increase using the conventional "BBBD protocol". This novel FUS approach offers a promising, cost-effective strategy for enhancing drug delivery to brain tumors. While further studies are required to assess its applicability with different chemotherapeutics and tumor types, it holds significant potential for improving brain tumor treatment in both preclinical and clinical settings.
Collapse
Affiliation(s)
- Hyo Jin Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Mun Han
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Byeongjin Jung
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
- BioHealth Convergence Center, Daegu Technopark, 46-17 Seongseogongdan-ro, Dalseo-gu, Daegu, 42716, Republic of Korea
| | - Hyungkyu Huh
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Eun-Hee Lee
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Jong-Ryul Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu, 41061, Republic of Korea
| | - Juyoung Park
- College of IT Convergence, Department of Biomedical Device, Gachon University, 1342, Seongnam-daero, Sujeong-gu, 1342, Seongnam, 13120, Gyeonggi, Republic of Korea.
- Neumous Inc., 1526, Seongnam-daero, Sujeong-gu, Seongnam-si, 13113, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
27
|
Zhao X, Li C, Wang K, Zhang Y, Wan H, Yang P, Zhou D. MR‑guided laser interstitial thermal therapy followed by early application of temozolomide for recurrent IDH-wildtype glioblastomas: preliminary results from a prospective study. Neurosurg Rev 2025; 48:254. [PMID: 39971799 DOI: 10.1007/s10143-025-03402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 01/19/2025] [Accepted: 02/09/2025] [Indexed: 02/21/2025]
Abstract
This study aims to evaluate the safety, tolerability, and preliminary efficacy of combining laser interstitial thermal therapy (LITT) with early administration of temozolomide (TMZ) in patients with recurrent glioblastoma (rGBM). Ten patients with rGBM were enrolled. Following the LITT procedure, TMZ was administered at a dose of 75 mg/m2/day during the early-TMZ phase for three weeks. After a 7-day interval, TMZ was given according to the standard dosage scheme for 6 cycles. Adverse events and complications encountered were documented. Regular follow-up assessments were conducted to evaluate both patient performance status and tumor progression. All patients demonstrated good tolerance to LITT, with six out of ten achieving an ablation rate above 90%, and only one patient had an ablation rate below 70%. Oral administration of TMZ was well-tolerated by all patients during the early-TMZ phase. Mild headache was the most common adverse event (3/10), and only one severe adverse event occurred. At a 6-month follow-up post-LITT, tumor progression was observed in five patients; noneof the patients reached survival endpoints. This preliminary report substantiates the favorable tolerability of early application of TMZ in combination with LITT. The safety profile was found to be acceptable, and the initial efficacy results were promising. Future studies should explore the potential of LITT combination therapy in greater detail and with larger patient samples.
Collapse
Affiliation(s)
- Xuzhe Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Chao Li
- Department of Neurosurgery, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Kai Wang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yutao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Haibin Wan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Pei Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China
| | - Dabiao Zhou
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
28
|
Liu Y, Wu H, Liang G. Combined Strategies for Nanodrugs Noninvasively Overcoming the Blood-Brain Barrier and Actively Targeting Glioma Lesions. Biomater Res 2025; 29:0133. [PMID: 39911305 PMCID: PMC11794768 DOI: 10.34133/bmr.0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 02/07/2025] Open
Abstract
Drugs for tumor treatment face various challenges, including poor solubility, poor stability, short blood half-life, nontargeting ability, and strong toxic side effects. Fortunately, nanodrug delivery systems provide excellent solution to these problems. However, nanodrugs for glioma treatment also face some key challenges including overcoming the blood-brain barrier (BBB) and, specifically, accumulation in glioma lesions. In this review, we systematically summarize the advantages and disadvantages of combined strategies for nanodrugs noninvasively overcoming BBB and actively targeting glioma lesions to achieve effective glioma therapy. Common noninvasive strategies for nanodrugs overcoming the BBB include bypassing the BBB via the nose-to-brain route, opening the tight junction of the BBB by focused ultrasound with microbubbles, and transendothelial cell transport by intact cell loading, ligand decoration, or cell membrane camouflage of nanodrugs. Actively targeting glioma lesions after overcoming the BBB is another key factor helping nanodrugs accurately treat in situ gliomas. This aim can also be achieved by loading nanodrugs into intact cells and modifying ligand or cell membrane fragments on the surface of nanodrugs. Targeting decorated nanodrugs can guarantee precise glioma killing and avoid side effects on normal brain tissues that contribute to the specific recognition of glioma lesions. Furthermore, the challenges and prospects of nanodrugs in clinical glioma treatment are discussed.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province 471000, China
| | - Haigang Wu
- Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan Province 475004, China
| | - Gaofeng Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, Henan Province 471000, China
| |
Collapse
|
29
|
Wei Y, Wang P, Zhao J, Fan X, Jiang J, Mu X, Wang Y, Yang A, Zhang R, Hu S, Guo Z. Overexpression of miR-124 enhances the therapeutic benefit of TMZ treatment in the orthotopic GBM mice model by inhibition of DNA damage repair. Cell Death Dis 2025; 16:47. [PMID: 39865088 PMCID: PMC11770086 DOI: 10.1038/s41419-025-07363-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: 08/31/2024] [Revised: 12/20/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain cancer with poor prognosis due to the resistant to current treatments, including the first-line drug temozolomide (TMZ). Accordingly, it is urgent to clarify the mechanism of chemotherapeutic resistance to improve the survival rate of patients. In the present study, by integrating comprehensive non-coding RNA-seq data from multiple cohorts of GBM patients, we identified that a series of miRNAs are frequently downregulated in GBM patients compared with the control samples. Among them, a high level of miR-124 is closely associated with a favorable survival rate in the clinical patients. In the phenotype experiment, we demonstrated that miR-124 overexpression increases responsiveness of GBM cells to TMZ-induced cell death, and vice versa. In the mechanistic study, we for the first time identified that RAD51, a key functional molecule in DNA damage repair, is a novel and bona fide target of miR-124 in GBM cells. Given that other miR-124-regulated mechanisms on TMZ sensitivity have been reported, we performed recue experiment to demonstrate that RAD51 is essential for miR-124-mediated sensitivity to TMZ in GBM cells. More importantly, our in vivo functional experiment showed that combinational utilization of miR-124 overexpression and TMZ presents a synergetic therapeutic benefit in the orthotopic GBM mice model. Taken together, we rationally explained a novel and important mechanism of the miR-124-mediated high sensitivity to TMZ-induced cell death in GBM and provided evidence to support that miR-124-RAD51 regulatory axis could be a promising candidate in the comprehensive treatment with TMZ in GBM.
Collapse
Affiliation(s)
- Yuchen Wei
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Peng Wang
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jianhui Zhao
- Department of Critical Care Medicine, Hainan Hospital of Chinese PLA General Hospital, Sanya City, Hainan Province, China
| | - Xin Fan
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jun Jiang
- Department of Health Service, Base of Health Service, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiuli Mu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yuzhou Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Angang Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Rui Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi Province, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China.
| | - Shijie Hu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China.
| | - Zhangyan Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
30
|
Rafanan J, Ghani N, Kazemeini S, Nadeem-Tariq A, Shih R, Vida TA. Modernizing Neuro-Oncology: The Impact of Imaging, Liquid Biopsies, and AI on Diagnosis and Treatment. Int J Mol Sci 2025; 26:917. [PMID: 39940686 PMCID: PMC11817476 DOI: 10.3390/ijms26030917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Advances in neuro-oncology have transformed the diagnosis and management of brain tumors, which are among the most challenging malignancies due to their high mortality rates and complex neurological effects. Despite advancements in surgery and chemoradiotherapy, the prognosis for glioblastoma multiforme (GBM) and brain metastases remains poor, underscoring the need for innovative diagnostic strategies. This review highlights recent advancements in imaging techniques, liquid biopsies, and artificial intelligence (AI) applications addressing current diagnostic challenges. Advanced imaging techniques, including diffusion tensor imaging (DTI) and magnetic resonance spectroscopy (MRS), improve the differentiation of tumor progression from treatment-related changes. Additionally, novel positron emission tomography (PET) radiotracers, such as 18F-fluoropivalate, 18F-fluoroethyltyrosine, and 18F-fluluciclovine, facilitate metabolic profiling of high-grade gliomas. Liquid biopsy, a minimally invasive technique, enables real-time monitoring of biomarkers such as circulating tumor DNA (ctDNA), extracellular vesicles (EVs), circulating tumor cells (CTCs), and tumor-educated platelets (TEPs), enhancing diagnostic precision. AI-driven algorithms, such as convolutional neural networks, integrate diagnostic tools to improve accuracy, reduce interobserver variability, and accelerate clinical decision-making. These innovations advance personalized neuro-oncological care, offering new opportunities to improve outcomes for patients with central nervous system tumors. We advocate for future research integrating these tools into clinical workflows, addressing accessibility challenges, and standardizing methodologies to ensure broad applicability in neuro-oncology.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Department of Medical Education, Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (J.R.); (N.G.); (S.K.); (A.N.-T.); (R.S.)
| |
Collapse
|
31
|
Szymulewska-Konopko K, Reszeć-Giełażyn J, Małeczek M. Ferritin as an Effective Prognostic Factor and Potential Cancer Biomarker. Curr Issues Mol Biol 2025; 47:60. [PMID: 39852175 PMCID: PMC11763953 DOI: 10.3390/cimb47010060] [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: 12/10/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
Ferritin is found in all cells of the body, serving as a reservoir of iron and protecting against damage to the molecules that make up cellular structures. It has emerged as a biomarker not only for iron-related disorders but also for inflammatory diseases and conditions in which inflammation plays a key role, including cancer, neurodegeneration, and infection. Oxidative stress, which can cause cellular damage, is induced by reactive oxygen species generated during the Fenton reaction, activating signaling pathways associated with tumor growth and proliferation. This review primarily emphasizes basic studies on the identification and function of ferritin, its essential role in iron metabolism, its involvement in inflammatory diseases, and its potential as an important prognostic factor and biomarker for cancer detection.
Collapse
Affiliation(s)
| | - Joanna Reszeć-Giełażyn
- Department of Medical Pathomorphology, Medical University of Bialystok, 15-089 Białystok, Poland; (K.S.-K.); (M.M.)
| | | |
Collapse
|
32
|
Ellingson BM, Okobi Q, Chong R, Plawat R, Zhao E, Gafita A, Sonni I, Chun S, Filka E, Yao J, Telesca D, Li S, Li G, Lai A, Nghiemphu P, Czernin J, Nathanson DA, Cloughesy TF. A comparative study of preclinical and clinical molecular imaging response to EGFR inhibition using osimertinib in glioblastoma. Neurooncol Adv 2025; 7:vdaf022. [PMID: 40051661 PMCID: PMC11883343 DOI: 10.1093/noajnl/vdaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
Background To demonstrate the potential value of 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) as a rapid, non-invasive metabolic imaging surrogate for pharmacological modulation of EGFR signaling in EGFR-driven GBM, we synchronously conducted a preclinical imaging study using patient-derived orthotopic xenograft (PDOX) models and validated it in a phase II molecular imaging study in recurrent GBM (rGBM) patients using osimertinib. Methods A GBM PDOX mouse model study was performed concurrently with an open-label, single-arm, single-center, phase II study of osimertinib (NCT03732352) that enrolled 12 patients with rGBM with EGFR alterations. Patients received osimertinib daily and 3 18F-FDG PET scans: two 24 h apart prior to dosing, and one 48 h after dosing. Results GBM PDOX models suggest osimertinib has limited impact on both 18F-FDG uptake (+ 9.8%-+25.9%) and survival (+ 15.5%; P = .01), which may be explained by insufficient exposure in the brain (Kpuu: 0.30) required to robustly inhibit the EGFR alterations found in GBM. Treatment with osimertinib had subtle, but measurable decreases in the linear rate of change of 18F-FDG nSUV growth rate averaging -4.5% per day (P = .01) and change in 18F-FDG uptake was correlated with change in tumor growth rate (R2 = 0.4719, P = .0195). No metabolic (PERCIST) or radiographic (RANO) responses were seen, and no improvements in PFS or OS were observed. Conclusions This study demonstrated the feasibility of using FDG PET as a clinically reliable imaging biomarker for assessing EGFR inhibition in GBM, while revealing osimertinib's limited impact on both metabolic activity and tumor growth in GBM, findings that were concordant between preclinical and clinical observations.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- UCLA Brain Tumor Imaging Laboratory (BTIL), Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Qunicy Okobi
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Robert Chong
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Rhea Plawat
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Eva Zhao
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Andrei Gafita
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Ida Sonni
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Saewon Chun
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Emese Filka
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Jingwen Yao
- UCLA Brain Tumor Imaging Laboratory (BTIL), Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Donatello Telesca
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Shanpeng Li
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Gang Li
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, California, USA
| | - Albert Lai
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Phioanh Nghiemphu
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
| | - Johannes Czernin
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - David A Nathanson
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Timothy F Cloughesy
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California, USA
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, USA
- Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
33
|
Giantini-Larsen AM, Pandey A, Garton ALA, Rampichini M, Winston G, Goldberg JL, Magge R, Stieg PE, Souweidane MM, Ramakrishna R. Therapeutic manipulation and bypass of the blood-brain barrier: powerful tools in glioma treatment. Neurooncol Adv 2025; 7:vdae201. [PMID: 39877748 PMCID: PMC11773386 DOI: 10.1093/noajnl/vdae201] [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: 01/31/2025] Open
Abstract
The blood-brain barrier (BBB) remains an obstacle for delivery of chemotherapeutic agents to gliomas. High grade and recurrent gliomas continue to portend a poor prognosis. Multiple methods of bypassing or manipulating the BBB have been explored, including hyperosmolar therapy, convection-enhanced delivery (CED), laser-guided interstitial thermal therapy (LITT), and Magnetic Resonance Guided Focused Ultrasound (MRgFUS) to enhance delivery of chemotherapeutic agents to glial neoplasms. Here, we review these techniques, currently ongoing clinical trials to disrupt or bypass the BBB in gliomas, and the results of completed trials.
Collapse
Affiliation(s)
- Alexandra M Giantini-Larsen
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Abhinav Pandey
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Andrew L A Garton
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Margherita Rampichini
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Graham Winston
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Jacob L Goldberg
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Rajiv Magge
- Department of Neurology, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Philip E Stieg
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Mark M Souweidane
- Department of Neurological Surgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY 10065, USA
| | - Rohan Ramakrishna
- Corresponding Author: Rohan Ramakrishna, MD, Chief, Neurological Surgery, New York Presbyterian Brooklyn Methodist Hospital, Weill Cornell Medical Center, 525 East 68 Street, New York, NY 10065, USA ()
| |
Collapse
|
34
|
Yu JS, Meade SM, Zhao R, Wei W, Dashora H, Prayson R, Grabowski MM, Stevens G, Lobbous M, Murphy ES, Suh JH, Chao ST, Barnett GH, Peereboom D, Ahluwalia MS, Mohammadi AM. Expedited chemoradiation after laser interstitial thermal therapy (LITT) is feasible and safe in patients with newly diagnosed glioblastoma. Neurooncol Adv 2025; 7:vdaf038. [PMID: 40276375 PMCID: PMC12019959 DOI: 10.1093/noajnl/vdaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025] Open
Abstract
Background High-grade gliomas (HGG) are incurable primary brain tumors. Laser interstitial thermal therapy (LITT) has emerged as an alternative to surgery for select patients. Hyperthermia can improve the efficacy of radiation and chemotherapy. Shortening the time between LITT and chemoradiation may maximize their biological and clinical benefits. This trial evaluated the safety and feasibility of expediting chemoradiation after biopsy and LITT in patients with newly diagnosed HGG. Methods Patients with suspected HGG were enrolled. Those with pathologic confirmation of HGG and deemed appropriate candidates for LITT and chemoradiation were considered evaluable. Participants underwent 6 weeks of adjuvant chemoradiation initiated within 7 days of LITT. Endpoints were assessed until the completion of radiation and included the occurrence of wound dehiscence; new, treatment-refractory seizures; cerebral edema; and completion of planned radiotherapy. Results Thirteen patients with suspected HGG were enrolled, and ten were considered evaluable. All 10 patients were diagnosed with glioblastoma (GBM, IDHwt). Three patients were deemed unevaluable: 2 patients with other CNS tumors and one GBM patient who developed grade 4 postoperative edema. Of 10 evaluable patients, the median age was 60.2 years (IQR: 51.0, 69.4), and median preoperative KPS was 90 (IQR: 90, 80). The median time between LITT and the initiation of chemoradiation was 7 days. There were no occurrences of significant protocol-related adverse events. Conclusions Accelerated initiation of chemoradiation after biopsy and LITT is safe and feasible for patients with newly diagnosed GBM. A larger study is needed to assess potential synergy of hyperthermia and chemoradiation to improve survival.
Collapse
Affiliation(s)
- Jennifer S Yu
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Seth M Meade
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ran Zhao
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Wei Wei
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Himanshu Dashora
- Case Western Reserve University School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richard Prayson
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Matthew M Grabowski
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Glen Stevens
- Department of Neurology, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mina Lobbous
- Department of Neurology, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Erin S Murphy
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - John H Suh
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Samuel T Chao
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Gene H Barnett
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - David Peereboom
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Manmeet S Ahluwalia
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alireza M Mohammadi
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio, USA
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
35
|
Ntafoulis I, Koolen SLW, van Tellingen O, den Hollander CWJ, Sabel-Goedknegt H, Dijkhuizen S, Haeck J, Reuvers TGA, de Bruijn P, van den Bosch TPP, van Dis V, Gao Z, Dirven CMF, Leenstra S, Lamfers MLM. A Repurposed Drug Selection Pipeline to Identify CNS-Penetrant Drug Candidates for Glioblastoma. Pharmaceuticals (Basel) 2024; 17:1687. [PMID: 39770529 PMCID: PMC11678797 DOI: 10.3390/ph17121687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Glioblastoma is an aggressive and incurable type of brain cancer. Little progress has been made in the development of effective new therapies in the past decades. The blood-brain barrier (BBB) and drug efflux pumps, which together hamper drug delivery to these tumors, play a pivotal role in the gap between promising preclinical findings and failure in clinical trials. Therefore, selecting drugs that can reach the tumor region in pharmacologically effective concentrations is of major importance. METHODS In the current study, we utilized a drug selection platform to identify candidate drugs by combining in vitro oncological drug screening data and pharmacokinetic (PK) profiles for central nervous system (CNS) penetration using the multiparameter optimization (MPO) score. Furthermore, we developed intracranial patient-derived xenograft (PDX) models that recapitulated the in situ characteristics of glioblastoma and characterized them in terms of vascular integrity, BBB permeability and expression of ATP-binding cassette (ABC) transporters. Omacetaxine mepesuccinate (OMA) was selected as a proof-of-concept drug candidate to validate our drug selection pipeline. RESULTS We assessed OMA's PK profile in three different orthotopic mouse PDX models and found that OMA reaches the brain tumor tissue at concentrations ranging from 2- to 11-fold higher than in vitro IC50 values on patient-derived glioblastoma cell cultures. CONCLUSIONS This study demonstrates that OMA, a drug selected for its in vitro anti-glioma activity and CNS- MPO score, achieves brain tumor tissue concentrations exceeding its in vitro IC50 values in patient-derived glioblastoma cell cultures, as shown in three orthotopic mouse PDX models. We emphasize the importance of such approaches at the preclinical level, highlighting both their significance and limitations in identifying compounds with potential clinical implementation in glioblastoma.
Collapse
Affiliation(s)
- Ioannis Ntafoulis
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands (C.M.F.D.); (S.L.)
| | - Stijn L. W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Chelsea W. J. den Hollander
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands (C.M.F.D.); (S.L.)
| | | | - Stephanie Dijkhuizen
- Department of Neuroscience, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands (Z.G.)
| | - Joost Haeck
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Thom G. A. Reuvers
- Department of Radiology & Nuclear Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Peter de Bruijn
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | | | - Vera van Dis
- Department of Pathology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (T.P.P.v.d.B.)
| | - Zhenyu Gao
- Department of Neuroscience, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands (Z.G.)
| | - Clemens M. F. Dirven
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands (C.M.F.D.); (S.L.)
| | - Sieger Leenstra
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands (C.M.F.D.); (S.L.)
| | - Martine L. M. Lamfers
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands (C.M.F.D.); (S.L.)
| |
Collapse
|
36
|
Liu Y, Zhou F, Ali H, Lathia JD, Chen P. Immunotherapy for glioblastoma: current state, challenges, and future perspectives. Cell Mol Immunol 2024; 21:1354-1375. [PMID: 39406966 PMCID: PMC11607068 DOI: 10.1038/s41423-024-01226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive and lethal type of brain tumor in human adults. The standard of care offers minimal clinical benefit, and most GBM patients experience tumor recurrence after treatment. In recent years, significant advancements have been made in the development of novel immunotherapies or other therapeutic strategies that can overcome immunotherapy resistance in many advanced cancers. However, the benefit of immune-based treatments in GBM is limited because of the unique brain immune profiles, GBM cell heterogeneity, and immunosuppressive tumor microenvironment. In this review, we present a detailed overview of current immunotherapeutic strategies and discuss the challenges and potential molecular mechanisms underlying immunotherapy resistance in GBM. Furthermore, we provide an in-depth discussion regarding the strategies that can overcome immunotherapy resistance in GBM, which will likely require combination therapies.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Fei Zhou
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Heba Ali
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
- Rose Ella Burkhardt Brain Tumor & Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA
| | - Peiwen Chen
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, 44195, USA.
| |
Collapse
|
37
|
Ghosh M, Roy D, Thakur S, Singh A. Exploring the Potential of Nasal Drug Delivery for Brain Targeted Therapy: A Detailed Analysis. Biopharm Drug Dispos 2024; 45:161-189. [PMID: 39665188 DOI: 10.1002/bdd.2400] [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: 08/21/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024]
Abstract
The brain is a sensitive organ with numerous essential functions and complex mechanisms. It is secluded and safeguarded from the external environment as part of the central nervous system (CNS), serving as a sanctuary. By regulating their selective and specific absorption, efflux, and metabolism in the brain, the CNS controls brain homeostasis and the transit of endogenous and foreign substances. The mechanism which protects the brain from environmental chemicals, also prevent the entry of therapeutic chemicals to it. The delivery of molecules to the brain is hindered by several major barriers, such as the blood-brain barrier (BBB), blood-cerebrospinal fluid barrier (BCSFB), and blood-tumor barrier. BBB is formed by the combination of cerebral endothelial cells, astrocytes, neurons, pericytes and microglia. It is a tight junction of capillary endothelial cells, preventing the diffusion of solute into the brain. BCSFB is the second barrier, located at the choroid plexus, separating the blood from cerebrospinal fluid (CSF). It is comparatively more permeable than BBB. An uneven distribution of microvasculature across the tumor interstitial compromises drug delivery to neoplastic cells of a solid tumor, resulting in spatially inconsistent drug administration. Nasal drug delivery to the brain is a method of drug delivery that tries to deliver therapeutic substances directly from the nasal cavity to the central nervous system including the brain. In this review, besides the role of barriers we have discussed in detail about approaches adapted to deliver drugs to the brain along with mechanisms through nasal route. Further, different commercial formulations, clinical trials and patents have been thoroughly elaborated to date. The findings suggest that the nose-to-brain drug delivery method holds promise as an evolving approach, potentially contributing to the specific and targeted delivery of drugs into the brain.
Collapse
Affiliation(s)
| | - Debajyoti Roy
- Department of Pharmacy, CV Raman Global University, Bhubaneswar, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, India
| | - Amrinder Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, India
| |
Collapse
|
38
|
You H, Geng S, Li S, Imani M, Brambilla D, Sun T, Jiang C. Recent advances in biomimetic strategies for the immunotherapy of glioblastoma. Biomaterials 2024; 311:122694. [PMID: 38959533 DOI: 10.1016/j.biomaterials.2024.122694] [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: 04/08/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Immunotherapy is regarded as one of the most promising approaches for treating tumors, with a multitude of immunotherapeutic thoughts currently under consideration for the lethal glioblastoma (GBM). However, issues with immunotherapeutic agents, such as limited in vivo stability, poor blood-brain barrier (BBB) penetration, insufficient GBM targeting, and represented monotherapy, have hindered the success of immunotherapeutic interventions. Moreover, even with the aid of conventional drug delivery systems, outcomes remain suboptimal. Biomimetic strategies seek to overcome these formidable drug delivery challenges by emulating nature's intelligent structures and functions. Leveraging the variety of biological structures and functions, biomimetic drug delivery systems afford a versatile platform with enhanced biocompatibility for the co-delivery of diverse immunotherapeutic agents. Moreover, their inherent capacity to traverse the BBB and home in on GBM holds promise for augmenting the efficacy of GBM immunotherapy. Thus, this review begins by revisiting the various thoughts and agents on immunotherapy for GBM. Then, the barriers to successful GBM immunotherapy are analyzed, and the corresponding biomimetic strategies are explored from the perspective of function and structure. Finally, the clinical translation's current state and prospects of biomimetic strategy are addressed. This review aspires to provide fresh perspectives on the advancement of immunotherapy for GBM.
Collapse
Affiliation(s)
- Haoyu You
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuo Geng
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shangkuo Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mohammad Imani
- Department of Science, Iran Polymer and Petrochemical Institute, Tehran 14977-13115, Iran; Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Tehran 14588-89694, Iran
| | - Davide Brambilla
- Faculty of Pharmacy, University of Montreal, Montreal Quebec H3T 1J4, Canada
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
39
|
Murphy KR, Aycock KN, Marsh S, Hay AN, Athanasiadi I, Bracha S, Chang C, Gourdie R, Davalos RV, Rossmeisl JH, Dervisis NG. Tumor-derived extracellular vesicles disrupt the blood-brain barrier endothelium following high-frequency irreversible electroporation. Sci Rep 2024; 14:28533. [PMID: 39557959 PMCID: PMC11574144 DOI: 10.1038/s41598-024-79019-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/05/2024] [Indexed: 11/20/2024] Open
Abstract
High-frequency irreversible electroporation (H-FIRE), a nonthermal brain tumor ablation therapeutic, generates a central tumor ablation zone while transiently disrupting the peritumoral blood-brain barrier (BBB). We hypothesized that bystander effects of H-FIRE tumor cell ablation, mediated by small tumor-derived extracellular vesicles (sTDEV), disrupt the BBB endothelium. Monolayers of bEnd.3 cerebral endothelial cells were exposed to supernatants of H-FIRE or radiation (RT)-treated LL/2 and F98 cancer cells. Endothelial cell response was evaluated microscopically and via flow cytometry for apoptosis. sTDEV were isolated following H-FIRE and RT, characterized via nanoparticle tracking analysis (NTA) and transmission electron microscopy, and applied to a Transwell BBB endothelium model to quantify permeability changes. Supernatants of H-FIRE-treated tumor cells, but not supernatants of sham- or RT-treated cells, disrupted endothelial cell monolayer integrity while maintaining viability. sTDEV released by glioma cells treated with 3000 V/cm H-FIRE increased permeability of the BBB endothelium model compared to sTDEV released after lower H-FIRE doses and RT. NTA revealed significantly decreased sTDEV release after the 3000 V/cm H-FIRE dose. Our results demonstrate that sTDEV increase permeability of the BBB endothelium after H-FIRE ablation in vitro. sTDEV-mediated mechanisms of BBB disruption may be exploited for drug delivery to infiltrative margins following H-FIRE ablation.
Collapse
Affiliation(s)
- Kelsey R Murphy
- Department of Biomedical and Veterinary Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA.
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA.
| | - Kenneth N Aycock
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
| | - Spencer Marsh
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA, USA
- Center for Heart and Reparative Medicine Research, Virginia Tech, Roanoke, VA, USA
| | - Alayna N Hay
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Ilektra Athanasiadi
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Shay Bracha
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Christine Chang
- Department of Veterinary Small Animal Clinical Sciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Robert Gourdie
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
- Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA, USA
- Center for Heart and Reparative Medicine Research, Virginia Tech, Roanoke, VA, USA
- Translational Biology Medicine and Health Graduate Program, Virginia Tech, Roanoke, VA, USA
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA, USA
| | - Rafael V Davalos
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA
- ICTAS Center for Engineered Health, Virginia Tech, Kelly Hall, Blacksburg, VA, USA
| | - John H Rossmeisl
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Nikolaos G Dervisis
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
40
|
Khuu A, Verreault M, Colin P, Tran H, Idbaih A. Clinical Applications of Antisense Oligonucleotides in Cancer: A Focus on Glioblastoma. Cells 2024; 13:1869. [PMID: 39594617 PMCID: PMC11592788 DOI: 10.3390/cells13221869] [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: 09/09/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Antisense oligonucleotides (ASOs) are promising drugs capable of modulating the protein expression of virtually any target with high specificity and high affinity through complementary base pairing. However, this requires a deep understanding of the target sequence and significant effort in designing the correct complementary drug. In addition, ASOs have been demonstrated to be well tolerated during their clinical use. Indeed, they are already used in many diseases due to pathogenic RNAs of known sequences and in several neurodegenerative diseases and metabolic diseases, for which they were given marketing authorizations (MAs) in Europe and the United States. Their use in oncology is gaining momentum with several identified targets, promising preclinical and clinical results, and recent market authorizations in the US. However, many challenges remain for their clinical use in cancer. It seems necessary to take a step back and review our knowledge of ASOs and their therapeutic uses in oncology. The objectives of this review are (i) to summarize the current state of the art of ASOs; (ii) to discuss the therapeutic use of ASOs in cancer; and (iii) to focus on ASO usage in glioblastoma, the challenges, and the perspective ahead.
Collapse
Affiliation(s)
- Alexandre Khuu
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Maïté Verreault
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| | - Philippe Colin
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Helene Tran
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Ahmed Idbaih
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| |
Collapse
|
41
|
Culkins C, Adomanis R, Phan N, Robinson B, Slaton E, Lothrop E, Chen Y, Kimmel BR. Unlocking the Gates: Therapeutic Agents for Noninvasive Drug Delivery Across the Blood-Brain Barrier. Mol Pharm 2024; 21:5430-5454. [PMID: 39324552 DOI: 10.1021/acs.molpharmaceut.4c00604] [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: 09/27/2024]
Abstract
The blood-brain barrier (BBB) is a highly selective network of various cell types that acts as a filter between the blood and the brain parenchyma. Because of this, the BBB remains a major obstacle for drug delivery to the central nervous system (CNS). In recent years, there has been a focus on developing various modifiable platforms, such as monoclonal antibodies (mAbs), nanobodies (Nbs), peptides, and nanoparticles, as both therapeutic agents and carriers for targeted drug delivery to treat brain cancers and diseases. Methods for bypassing the BBB can be invasive or noninvasive. Invasive techniques, such as transient disruption of the BBB using low pulse electrical fields and intracerebroventricular infusion, lack specificity and have numerous safety concerns. In this review, we will focus on noninvasive transport mechanisms that offer high levels of biocompatibility, personalization, specificity and are regarded as generally safer than their invasive counterparts. Modifiable platforms can be designed to noninvasively traverse the BBB through one or more of the following pathways: passive diffusion through a physio-pathologically disrupted BBB, adsorptive-mediated transcytosis, receptor-mediated transcytosis, shuttle-mediated transcytosis, and somatic gene transfer. Through understanding the noninvasive pathways, new applications, including Chimeric Antigen Receptors T-cell (CAR-T) therapy, and approaches for drug delivery across the BBB are emerging.
Collapse
Affiliation(s)
- Courtney Culkins
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Roman Adomanis
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nathan Phan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise Robinson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ethan Slaton
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Elijah Lothrop
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yinuo Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise R Kimmel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Engineering, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
42
|
Singh S, Bhardwaj S, Dandapath I, Singh J, Das S, Mohan T, Bora SK, Kedia S, Suri A, Sharma MC, Sarkar C, Faruq M, Suri V. Clinical utility of plasma cell-free DNA (cfDNA) in diffuse gliomas for the detection of IDH1 R132H mutation. Pathol Res Pract 2024; 263:155571. [PMID: 39298928 DOI: 10.1016/j.prp.2024.155571] [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: 06/30/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
Liquid biopsy for CNS tumors is in its nascent phase, hindered by the low levels of circulating tumor DNA (ctDNA). Overcoming this challenge requires highly sensitive molecular techniques. DD-PCR emerges as a standout technique due to its ability to identify rare mutations, copy number variations, and circulating nucleic acids, making it one of the best methods for identifying somatic mutations in cell-free DNA (cfDNA). Despite promising results from various studies demonstrating the feasibility of obtaining informative ctDNA profiles from liquid biopsy samples, challenges persist, including the need to standardize sample collection, storage, and processing methods, define clear assay positivity thresholds, and address the overall low assay sensitivity. Our two-phase study began by assessing DD-PCR efficacy in FFPE tissues, revealing robust concordance with immunohistochemistry. In Phase 1 (85 cases), DD-PCR on FFPE tissues demonstrated 100 % sensitivity and specificity for IDH1 R132H mutations. In Phase 2 (100 cases), analysis extended to cfDNA, maintaining high specificity (100 %) with moderate sensitivity (44.2 %). Overall concordance with immunohistochemistry was 61 %, highlighting liquid biopsy's potential in glioma management. The findings emphasized DD-PCR's clinical utility in both tissue and liquid biopsy, underscoring its role in early detection, diagnosis, and therapeutic monitoring of diffuse gliomas.
Collapse
Affiliation(s)
- Swati Singh
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Supriya Bhardwaj
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Iman Dandapath
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Jyotsna Singh
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Sumanta Das
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Trishala Mohan
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Santanu Kumar Bora
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Shweta Kedia
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Suri
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Mehar Chand Sharma
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Mohammed Faruq
- CSIR Institute of Genomics and Integrative Biology, Delhi, India
| | - Vaishali Suri
- Neuropathology Laboratory, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
43
|
Dragojevic S, Smith EJ, Regan MS, Stopka SA, Baquer G, Xue Z, Zhang W, Connors MA, Kloeber JA, Hu Z, Bakken KK, Ott LL, Carlson BL, Burgenske DM, Decker PA, Tian S, Gupta SK, Laverty DJ, Eckel-Passow JE, Elmquist WF, Agar NY, Nagel ZD, Sarkaria JN, Callaghan CM. DNA-PK inhibition shows differential radiosensitization in orthotopic GBM PDX models based on DDR pathway deficits. Mol Cancer Ther 2024:749287. [PMID: 39440433 PMCID: PMC12014860 DOI: 10.1158/1535-7163.mct-24-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/06/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024]
Abstract
Glioblastoma (GBM) remains one of the most therapy-resistant malignancies with frequent local failures despite aggressive surgery, chemotherapy, and ionizing radiation (IR). Small molecule inhibitors of DNA-dependent protein kinase (DNA-PKi's) are potent radiosensitizers currently in clinical trials. Determining which patients may benefit from radiosensitization with DNA-PKi's is critical to avoid unnecessary increased risk of normal tissue toxicity. In this study we used GBM patient derived xenografts (PDXs) in orthotopic murine models to study the relationship between molecular features, pharmacokinetics, and the radiosensitizing potential of the DNA-PKi peposertib. We show that peposertib radiosensitizes established and PDX GBM lines in vitro at 300nM and above, with significant increase in radiosensitization by maintaining post-IR exposure for >12 hours. Radiosensitization by peposertib is mediated by catalytic inhibition of DNA-PK, and knock-down of DNA-PK by short hairpin RNA (shRNA) largely abolished the radiosensitizing effect. Peposertib decreased auto-phosphorylation of DNA-PKcs after IR in a dose-dependent manner with delay in resolution of γH2AX foci at 24 hours. The addition of peposertib to IR significantly increased survival in GBM120 orthotopic xenografts, but not in GBM10. There was no difference in plasma or average tumor concentrations of peposertib in the two cohorts. While the mechanism underpinning this discordant effect in vitro vs. in vivo is not clear, there was an association for greater sensitization in TP53 mutant lines. Transfection of a dominant-negative TP53 mutant in baseline TP53 wildtype GBM lines significantly delayed growth and decreased NHEJ efficiency (but not Homologous Recombination), after peposertib exposure.
Collapse
Affiliation(s)
- Sonja Dragojevic
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Emily J. Smith
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael S. Regan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sylwia A. Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gerard Baquer
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhiyi Xue
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Wenjuan Zhang
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Jake A. Kloeber
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN, USA
| | - Zeng Hu
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Katrina K. Bakken
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lauren L. Ott
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brett L. Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Paul A. Decker
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Shulan Tian
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Shiv K. Gupta
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - William F. Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nathalie Y.R. Agar
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zachary D. Nagel
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
44
|
Rathi S, Mladek AC, Oh JH, Dragojevic S, Burgenske DM, Zhang W, Talele S, Zhang W, Bakken KK, Carlson BL, Connors MA, He L, Hu Z, Sarkaria JN, Elmquist WF. Factors Influencing the Central Nervous System (CNS) Distribution of the Ataxia Telangiectasia Mutated and Rad3-Related Inhibitor Elimusertib (BAY1895344): Implications for the Treatment of CNS Tumors. J Pharmacol Exp Ther 2024; 391:346-360. [PMID: 39284626 PMCID: PMC11493447 DOI: 10.1124/jpet.123.002002] [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: 11/01/2023] [Accepted: 09/09/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is a disease of the whole brain, with infiltrative tumor cells protected by an intact blood-brain barrier (BBB). GBM has a poor prognosis despite aggressive treatment, in part due to the lack of adequate drug permeability at the BBB. Standard of care GBM therapies include radiation and cytotoxic chemotherapy that lead to DNA damage. Subsequent activation of DNA damage response (DDR) pathways can induce resistance. Various DDR inhibitors, targeting the key regulators of these pathways such as ataxia telangiectasia mutated and Rad3-related (ATR), are being explored as radio- and chemosensitizers. Elimusertib, a novel ATR kinase inhibitor, can prevent repair of damaged DNA, increasing efficacy of DNA-damaging cytotoxic therapies. Robust synergy was observed in vitro when elimusertib was combined with the DNA-damaging agent temozolomide; however, we did not observe improvement with this combination in in vivo efficacy studies in GBM orthotopic tumor-bearing mice. This in vitro-in vivo disconnect was explored to understand factors influencing central nervous system (CNS) distribution of elimusertib and reasons for lack of efficacy. We observed that elimusertib is rapidly cleared from systemic circulation in mice and would not maintain adequate exposure in the CNS for efficacious combination therapy with temozolomide. CNS distribution of elimusertib is partially limited by P-glycoprotein efflux at the BBB, and high binding to CNS tissues leads to low levels of pharmacologically active (unbound) drug in the brain. Acknowledging the potential for interspecies differences in pharmacokinetics, these data suggest that clinical translation of elimusertib in combination with temozolomide for treatment of GBM may be limited. SIGNIFICANCE STATEMENT: This study examined the disconnect between the in vitro synergy and in vivo efficacy of elimusertib/temozolomide combination therapy by exploring systemic and central nervous system (CNS) distributional pharmacokinetics. Results indicate that the lack of improvement in in vivo efficacy in glioblastoma (GBM) patient-derived xenograft (PDX) models could be attributed to inadequate exposure of pharmacologically active drug concentrations in the CNS. These observations can guide further exploration of elimusertib for the treatment of GBM or other CNS tumors.
Collapse
Affiliation(s)
- Sneha Rathi
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Ann C Mladek
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Ju-Hee Oh
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Sonja Dragojevic
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Danielle M Burgenske
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Wenjuan Zhang
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Surabhi Talele
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Wenqiu Zhang
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Katrina K Bakken
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Brett L Carlson
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Margaret A Connors
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Lihong He
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Zeng Hu
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - Jann N Sarkaria
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (S.R., J.-H.O., W.J.Z., S.T., W.Q.Z., W.F.E.) and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (A.C.M., S.D., D.M.B., K.K.B., B.L.C., M.A.C., L.H., Z.H., J.N.S.)
| |
Collapse
|
45
|
Yan RE, Greenfield JP. Challenges and Outlooks in Precision Medicine: Expectations Versus Reality. World Neurosurg 2024; 190:573-581. [PMID: 39425299 DOI: 10.1016/j.wneu.2024.06.142] [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: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 10/21/2024]
Abstract
Recent developments in technology have led to rapid advances in precision medicine, especially due to the rise of next-generation sequencing and molecular profiling. These technological advances have led to rapid advances in research, including increased tumor subtype resolution, new therapeutic agents, and mechanistic insights. Certain therapies have even been approved for molecular biomarkers across histopathological diagnoses; however, translation of research findings to the clinic still faces a number of challenges. In this review, the authors discuss several key challenges to the clinical integration of precision medicine, including the blood-brain barrier, both a lack and excess of molecular targets, and tumor heterogeneity/escape from therapy. They also highlight a few key efforts to address these challenges, including new frontiers in drug delivery, a rapidly expanding treatment repertoire, and improvements in active response monitoring. With continued improvements and developments, the authors anticipate that precision medicine will increasingly become the gold standard for clinical care.
Collapse
Affiliation(s)
- Rachel E Yan
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Jeffrey P Greenfield
- Department of Neurological Surgery, NewYork-Presbyterian Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
46
|
Mao M, Yang W, Zhang X. Current mRNA-based vaccine strategies for glioma treatment. Crit Rev Oncol Hematol 2024; 202:104459. [PMID: 39097247 DOI: 10.1016/j.critrevonc.2024.104459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/05/2024] Open
Abstract
Gliomas are one of the most aggressive types of brain tumors and are associated with high morbidity and mortality rates. Currently, conventional treatments for gliomas such as surgical resection, radiotherapy, and chemotherapy have limited effectiveness, and new approaches are needed to improve patient outcomes. mRNA-based vaccines represent a promising therapeutic strategy for cancer treatment, including gliomas. Recent advances in immunotherapy using mRNA-based dendritic cell vaccines have shown great potential in preclinical and clinical trials. Dendritic cells are professional antigen-presenting cells that play a crucial role in initiating and regulating immune responses. In this review, we summarize the current progress of mRNA-based vaccines for gliomas, with a focus on recent advances in dendritic cell-based mRNA vaccines. We also discuss the feasibility and safety of mRNA-based clinical applications for gliomas.
Collapse
Affiliation(s)
- Mengqian Mao
- Neuroscience & Metabolism Research, Department of Neurosurgery, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Wanchun Yang
- Neuroscience & Metabolism Research, Department of Neurosurgery, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Xuefeng Zhang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, China.
| |
Collapse
|
47
|
Hautiere M, Vivier D, Dorval P, Pineau D, Kereselidze D, Denis C, Herbet A, Costa N, Bernhard C, Goncalves V, Selingue E, Larrat B, Dancer PA, Hugnot JP, Boquet D, Truillet C, Denat F. Preoperative PET imaging and fluorescence-guided surgery of human glioblastoma using dual-labeled antibody targeting ET A receptors in a preclinical mouse model: A theranostic approach. Theranostics 2024; 14:6268-6280. [PMID: 39431005 PMCID: PMC11488107 DOI: 10.7150/thno.98163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/08/2024] [Indexed: 10/22/2024] Open
Abstract
Rationale: Glioblastoma (GBM) poses significant challenges regarding complete tumor removal due to its heterogeneity and invasiveness, emphasizing the need for effective therapeutic options. In the last two decades, fluorescence-guided surgery (FGS), employing fluorophores such as 5-aminolevulinic acid (5-ALA) to enhance tumor delineation, has gained attraction among neurosurgeons. However, some low-grade tumors do not show any accumulation of the tracers, and the lack of patient stratification represents an important limitation. Since 2000, endothelin axis has been extensively investigated for its role in cancer progression. More specifically, our team has identified endothelin A receptors (ETA), overexpressed in glioblastoma cancer stem cells, as a target of interest for GBM imaging. This study aims to evaluate the efficacy of a novel preclinical bimodal imaging agent, [89Zr]Zr-axiRA63-MOMIP, as a theranostic approach to: i) detect ETA + cells in an orthotopic model of human GBM, ii) achieve complete tumoral resection. Methods: Monomolecular multimodal imaging platform (MOMIP) - containing both a fluorophore (IRDye800CW) and a chelator for a positron-emitting radiometal (desferroxamine B, DFO) - was conjugated to the axiRA63 antibody targeting ETA receptors, overexpressed on the surface of GBM stem cells. Mice bearing orthotopic human GBM were imaged 48 h post injection of [89Zr]Zr-axiRA63-MOMIP via positron emission tomography (PET) and optical imaging. Subsequently, post-mortem proof-of-concept FGS was implemented as well as ex vivo analyses (H&E staining, autoradiography, serial block face imaging) on brains with resected or unresected tumor to assess the correlation between PET and fluorescence signals. Results: PET imaging of [89Zr]Zr-axiRA63-MOMIP enabled a clear detection of ETA + cells in an orthotopic model of human GBM. Intraoperative optical imaging allowed a near-complete tumor resection together with the visualization of a weak fluorescence signal, after a prolonged exposure time, that was attributed to residual tumor cells via H&E staining. Besides, a qualitative correlation between the signals of both modalities was observed. Conclusions: The use of [89Zr]Zr-axiRA63-MOMIP provides an effective theranostic approach to detect and treat GBM by surgery in a preclinical mouse model. Thanks to the high correlation between PET and fluorescence signal allowing patients stratification, this bimodal agent should have a great potential for clinical translation and should present a significant advantage over non-targeted fluorophores already used in the clinic.
Collapse
Affiliation(s)
- Marie Hautiere
- Université Paris-Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (MTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire et Innovation Thérapeutique (LENIT), 91191 Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay, 91401, France
| | - Delphine Vivier
- Université de Bourgogne, ICMUB UMR CNRS 6302, Dijon, 21000, France
| | | | - Donovan Pineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Caroline Denis
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay, 91401, France
| | - Amaury Herbet
- Université Paris-Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (MTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire et Innovation Thérapeutique (LENIT), 91191 Gif-sur-Yvette, France
| | - Narciso Costa
- Université Paris-Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (MTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire et Innovation Thérapeutique (LENIT), 91191 Gif-sur-Yvette, France
| | - Claire Bernhard
- Université de Bourgogne, ICMUB UMR CNRS 6302, Dijon, 21000, France
| | - Victor Goncalves
- Université de Bourgogne, ICMUB UMR CNRS 6302, Dijon, 21000, France
| | - Erwan Selingue
- Université Paris-Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Gif sur Yvette, France
| | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Gif sur Yvette, France
| | | | - Jean-Philippe Hugnot
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Didier Boquet
- Université Paris-Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (MTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire et Innovation Thérapeutique (LENIT), 91191 Gif-sur-Yvette, France
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay, 91401, France
| | - Franck Denat
- Université de Bourgogne, ICMUB UMR CNRS 6302, Dijon, 21000, France
| |
Collapse
|
48
|
Sipos D, Debreczeni-Máté Z, Ritter Z, Freihat O, Simon M, Kovács Á. Complex Diagnostic Challenges in Glioblastoma: The Role of 18F-FDOPA PET Imaging. Pharmaceuticals (Basel) 2024; 17:1215. [PMID: 39338377 PMCID: PMC11434841 DOI: 10.3390/ph17091215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Glioblastoma multiforme (GBM) remains one of the most aggressive and lethal forms of brain cancer, characterized by rapid proliferation and diffuse infiltration into the surrounding brain tissues. Despite advancements in therapeutic approaches, the prognosis for GBM patients is poor, with median survival times rarely exceeding 15 months post-diagnosis. An accurate diagnosis, treatment planning, and monitoring are crucial for improving patient outcomes. Core imaging modalities such as Computed Tomography (CT) and Magnetic Resonance Imaging (MRI) are indispensable in the initial diagnosis and ongoing management of GBM. Histopathology remains the gold standard for definitive diagnoses, guiding treatment by providing molecular and genetic insights into the tumor. Advanced imaging modalities, particularly positron emission tomography (PET), play a pivotal role in the management of GBM. Among these, 3,4-dihydroxy-6-[18F]-fluoro-L-phenylalanine (18F-FDOPA) PET has emerged as a powerful tool due to its superior specificity and sensitivity in detecting GBM and monitoring treatment responses. This introduction provides a comprehensive overview of the multifaceted role of 18F-FDOPA PET in GBM, covering its diagnostic accuracy, potential as a biomarker, integration into clinical workflows, impact on patient outcomes, technological and methodological advancements, comparative effectiveness with other PET tracers, and its cost-effectiveness in clinical practice. Through these perspectives, we aim to underscore the significant contributions of 18F-FDOPA PET to the evolving landscape of GBM management and its potential to enhance both clinical and economic outcomes for patients afflicted with this formidable disease.
Collapse
Affiliation(s)
- David Sipos
- Department of Medical Imaging, Faculty of Health Sciences, University of Pécs, 7621 Pécs, Hungary
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, 7621 Pécs, Hungary
- Dr. József Baka Diagnostic, Radiation Oncology, Research and Teaching Center, “Moritz Kaposi” Teaching Hospital, Guba Sándor Street 40, 7400 Kaposvár, Hungary
| | - Zsanett Debreczeni-Máté
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, 7621 Pécs, Hungary
| | - Zsombor Ritter
- Department of Medical Imaging, Medical School, University of Pécs, 7621 Pécs, Hungary
| | - Omar Freihat
- Department of Public Health, College of Health Science, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Mihály Simon
- Department of Oncoradiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Árpád Kovács
- Department of Medical Imaging, Faculty of Health Sciences, University of Pécs, 7621 Pécs, Hungary
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, 7621 Pécs, Hungary
- Department of Oncoradiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
49
|
Ma C, Li X, Mao N, Wang M, Cui C, Jia H, Liu X, Sun Q. Semi-invasive wearable clinic: Solution-processed smart microneedle electronics for next-generation integrated diagnosis and treatment. Biosens Bioelectron 2024; 260:116427. [PMID: 38823368 DOI: 10.1016/j.bios.2024.116427] [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: 02/23/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 06/03/2024]
Abstract
The integrated smart electronics for real-time monitoring and personalized therapy of disease-related analytes have been gradually gaining tremendous attention. However, human tissue barriers, including the skin barrier and brain-blood barrier, pose significant challenges for effective biomarker detection and drug delivery. Microneedle (MN) electronics present a promising solution to overcome these tissue barriers due to their semi-invasive structures, enabling effective drug delivery and target-analyte detection without compromising the tissue configuration. Furthermore, MNs can be fabricated through solution processing, facilitating large-scale manufacturing. This review provides a comprehensive summary of the recent three-year advancements in smart MNs development, categorized as follows. First, the solution-processed technology for MNs is introduced, with a focus on various printing technologies. Subsequently, smart MNs designed for sensing, drug delivery, and integrated systems combining diagnosis and treatment are separately summarized. Finally, the prospective and promising applications of next-generation MNs within mediated diagnosis and treatment systems are discussed.
Collapse
Affiliation(s)
- Chao Ma
- School of Materials Science and Engineering, Zhengzhou Key Laboratory of Flexible Electronic Materials and Thin-Film Technologies, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- National Center for International Joint Research of Micro-nano Molding Technology, School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Ning Mao
- School of Materials Science and Engineering, Zhengzhou Key Laboratory of Flexible Electronic Materials and Thin-Film Technologies, Zhengzhou University, Zhengzhou 450001, China
| | - Mengwei Wang
- School of Materials Science and Engineering, Zhengzhou Key Laboratory of Flexible Electronic Materials and Thin-Film Technologies, Zhengzhou University, Zhengzhou 450001, China
| | - Cancan Cui
- School of Materials Science and Engineering, Zhengzhou Key Laboratory of Flexible Electronic Materials and Thin-Film Technologies, Zhengzhou University, Zhengzhou 450001, China
| | - Hanyu Jia
- School of Materials Science and Engineering, Zhengzhou Key Laboratory of Flexible Electronic Materials and Thin-Film Technologies, Zhengzhou University, Zhengzhou 450001, China
| | - Xuying Liu
- School of Materials Science and Engineering, Zhengzhou Key Laboratory of Flexible Electronic Materials and Thin-Film Technologies, Zhengzhou University, Zhengzhou 450001, China
| | - Qingqing Sun
- School of Materials Science and Engineering, Zhengzhou Key Laboratory of Flexible Electronic Materials and Thin-Film Technologies, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
50
|
Pfeil AJ, Hale JD, Zhang TS, Wakayama K, Miyazaki I, Odintsov I, Somwar R. Preclinical evaluation of targeted therapies for central nervous system metastases. Dis Model Mech 2024; 17:dmm050836. [PMID: 39344915 PMCID: PMC11463968 DOI: 10.1242/dmm.050836] [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: 10/01/2024] Open
Abstract
The central nervous system (CNS) represents a site of sanctuary for many metastatic tumors when systemic therapies that control the primary tumor cannot effectively penetrate intracranial lesions. Non-small cell lung cancers (NSCLCs) are the most likely of all neoplasms to metastasize to the brain, with up to 60% of patients developing CNS metastases during the disease process. Targeted therapies such as tyrosine kinase inhibitors (TKIs) have helped reduce lung cancer mortality but vary considerably in their capacity to control CNS metastases. The ability of these therapies to effectively target lesions in the CNS depends on several of their pharmacokinetic properties, including blood-brain barrier permeability, affinity for efflux transporters, and binding affinity for both plasma and brain tissue. Despite the existence of numerous preclinical models with which to characterize these properties, many targeted therapies have not been rigorously tested for CNS penetration during the discovery process, whereas some made it through preclinical testing despite poor brain penetration kinetics. Several TKIs have now been engineered with the characteristics of CNS-penetrant drugs, with clinical trials proving these efforts fruitful. This Review outlines the extent and variability of preclinical evidence for the efficacy of NSCLC-targeted therapies, which have been approved by the US Food and Drug Administration (FDA) or are in development, for treating CNS metastases, and how these data correlate with clinical outcomes.
Collapse
Affiliation(s)
- Alexander J. Pfeil
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Joshua D. Hale
- University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Tiger S. Zhang
- University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Kentaro Wakayama
- Taiho Pharmaceutical Co. Ltd. 3, Okubo, Tsukuba, Ibaraki 300-2611, Japan
| | - Isao Miyazaki
- Taiho Pharmaceutical Co. Ltd. 3, Okubo, Tsukuba, Ibaraki 300-2611, Japan
| | - Igor Odintsov
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 021105, USA
| | - Romel Somwar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|