1
|
Zhang W, Wang J, Ji J, Wang P, Yuan G, Fang S, Liu F, Jin G, Zhang J. Glioblastoma cells secrete ICAM1 via FASN signaling to promote glioma-associated macrophage infiltration. Cell Signal 2025; 132:111823. [PMID: 40252818 DOI: 10.1016/j.cellsig.2025.111823] [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: 01/30/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Glioma-associated macrophages (GAMs) constitute the most abundant subset of immune cells in the glioblastoma (GBM) microenvironment, but the underlying mechanism of intense infiltration needs to be elucidated. In this study, we found that GBM cells secrete ICAM1 via FASN signaling to promote GAM infiltration. FASN expression is correlated with GAM density in GBM patients. In vitro experiments revealed that FASN regulates the type-I interferon pathway, particularly STAT1 expression. Moreover, disrupting FASN-STAT1 signaling through the overexpression or inhibition of FASN or STAT1 in GBM cells strongly influences microglial recruitment. Additionally, ICAM1 acts as a direct transcriptional candidate of FASN-STAT1 and a paracrine soluble factor, recruiting microglia to GBM tumors. This study revealed crosstalk between GBM cells and GAMs through FASN-STAT1-ICAM1 signaling to promote microglial infiltration, suggesting potential strategies for treating GBM patients.
Collapse
Affiliation(s)
- Wenxin Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jialin Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiayu Ji
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China
| | - Peiwen Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Guiqiang Yuan
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Department of Neurosurgery, Beijing Tiantan Hosipital, Capital Medical University, Beijing, China
| | - Guishan Jin
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
3
|
Chen Z, Sang L, Qixi Z, Li X, Liu Y, Bai Z. Ultrasound-responsive nanoparticles for imaging and therapy of brain tumors. Mater Today Bio 2025; 32:101661. [PMID: 40206140 PMCID: PMC11979416 DOI: 10.1016/j.mtbio.2025.101661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/26/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
Central nervous system (CNS) cancers, particularly glioblastoma (GBM), are associated with high mortality and disability rates. Despite aggressive surgical resection, radiotherapy, and chemotherapy, patient survival remains poor. The blood-brain barrier (BBB) significantly impedes therapeutic efficacy, making BBB penetration a critical focus of research. Focused ultrasound (FUS) combined with microbubbles (MBs) can transiently open the BBB through mechanisms such as cavitation, modulation of tight junction protein expression, and enhanced vesicular transport in endothelial cells. This review highlights precision delivery and personalized treatment strategies under ultrasound visualization, including precise control of ultrasound parameters and modulation of the immune microenvironment. We discuss the applications of ultrasound-responsive nanoparticles in brain tumor therapy, including enhanced radiotherapy, gene delivery, immunotherapy, and sonodynamic therapy (SDT), with a particular emphasis on piezoelectric catalytic immunotherapy. Finally, we provide insights into the clinical translation potential of ultrasound-responsive nanoparticles for personalized and precision treatment of brain tumors.
Collapse
Affiliation(s)
- Zhiguang Chen
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Liang Sang
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Zhai Qixi
- Department of Ultrasound, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | | | | | | |
Collapse
|
4
|
Yamaguchi H, Okada M, Otani T, On J, Shibuma S, Takino T, Watanabe J, Tsukamoto Y, Ogura R, Oishi M, Suzuki T, Ishikawa A, Sakata H, Natsumeda M. Near-Infrared Photoimmunotherapy in Brain Tumors-An Unexplored Frontier. Pharmaceuticals (Basel) 2025; 18:751. [PMID: 40430568 DOI: 10.3390/ph18050751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/16/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a promising cancer treatment that uses near-infrared light to activate a conjugate of a monoclonal antibody (mAb) and a photoactivatable silica phthalocyanine dye (IRDye700DX: IR700). Unlike conventional photodynamic therapy (PDT), NIR-PIT selectively destroys targeted tumor cells while preserving the surrounding normal tissue and providing superior tissue penetration. Recently, NIR-PIT has been approved for the treatment of unresectable recurrent head and neck cancers in Japan. It induces highly selective cancer cell death; therefore, it is expected to be a new curative treatment option for various cancers, including brain tumors. In this review, we compare the principles of NIR-PIT and PDT and discuss the potential applications of NIR-PIT for brain tumors. We selected targetable proteins across various types of brain tumors and devised a strategy to effectively pass the mAb-IR700 conjugate through the blood-brain barrier (BBB), which is a significant challenge for NIR-PIT in treating brain tumors. Innovative approaches for delivering the mAb-IR700 conjugate across the BBB include exosomes, nanoparticle-based systems, and cell-penetrating peptides. Small-molecule compounds, such as affibodies, are anticipated to rapidly accumulate in tumors within intracranial models, and our preliminary experiments demonstrated rapid uptake. NIR-PIT also induces immunogenic cell death and activates the anti-tumor immune response. Overall, NIR-PIT is a promising approach for treating brain tumors. It has the potential to overcome the limitations of conventional therapies and offers new hope to patients with brain tumors.
Collapse
Affiliation(s)
- Haruka Yamaguchi
- Department of Biochemistry, School of Life Dentistry at Niigata, The Nippon Dental University, Niigata 951-8580, Japan
| | - Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Takuya Otani
- Near InfraRed Photo-ImmunoTherapy Research Institute, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Jotaro On
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Satoshi Shibuma
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Toru Takino
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Jun Watanabe
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Yoshihiro Tsukamoto
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Ryosuke Ogura
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Makoto Oishi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Takamasa Suzuki
- Faculty of Engineering, Niigata University, Niigata 950-2181, Japan
| | - Akihiro Ishikawa
- Startup Incubation Center, Shimadzu Corporation, Kyoto 604-8511, Japan
| | - Hideyuki Sakata
- Startup Incubation Center, Shimadzu Corporation, Kyoto 604-8511, Japan
| | - Manabu Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
- Advanced Treatment of Neurological Diseases Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| |
Collapse
|
5
|
Song J, Lee Y, Kim MS, Ha G, Jang W, Batjargal U, Kim Y, Kim HJ, Lee J. High throughput drug screening platform utilizing capillary and artery cell layered models based on tumor-vascular cell interactions. LAB ON A CHIP 2025; 25:2349-2363. [PMID: 40177711 DOI: 10.1039/d4lc00950a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Interactions between tumors and adjacent blood vessels are critical in the tumor microenvironment (TME) for influencing angiogenesis and hematogenous metastasis. Understanding these interactions within the native TME is vital for targeting various tumors, including brain tumors, due to the complexities of the blood-brain barrier. Developing an accurate tumor model that includes cell-cell and cell-matrix interactions, as well as blood flow-induced shear stress, is essential for high-throughput screening (HTS) of anti-cancer drugs. Here, we developed a glioblastoma (GBM) model surrounded by vascular cells. The arterial model was constructed by encapsulating GBM spheroids with layers of human smooth muscle cells (SMCs) and human umbilical vein endothelial cells (HUVECs), while the capillary cell layered model used only HUVECs. Comparative analysis with tumors from different organs revealed the significant role for platelet endothelial cell adhesion molecule (PECAM) in GBM-blood vascular cell interactions. Cytokine secretion analysis demonstrated PECAM's impact on tumor-specific angiogenic potential. Testing with anti-cancer drugs revealed increased expression of PECAM-associated proteins, drug resistance cytokines, and genes associated with tumor progression and metastasis. Additionally, we developed a HTS platform by encapsulating these tumor models in hydrogels and subjecting them to media circulation, effectively mimicking the dynamic TME, suitable for cancer treatment research and drug development.
Collapse
Affiliation(s)
- Jihyeon Song
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea.
| | - Yeji Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea.
| | - Min-Seok Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea.
| | - Giheon Ha
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea.
| | - WonJun Jang
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea.
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Ulziituya Batjargal
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea.
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Younggyun Kim
- Department of Bioengineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Han-Jun Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea.
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea.
| |
Collapse
|
6
|
Goo J, Lee JS, Park J, Jeon SI, Kim J, Yun WS, Shim N, Moon Y, Song S, Kim J, Cho H, Jeong JY, Lee JS, Han S, Lee HJ, Koh WG, Chang WS, Kim TI, Kim K. Dual Delivery of Light/Prodrug Nanoparticles Using Tumor-Implantable Micro Light-Emitting Diode on an Optofluidic System for Combinational Glioma Treatment. ACS NANO 2025; 19:17393-17409. [PMID: 40296430 DOI: 10.1021/acsnano.4c17421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Glioma is a highly lethal tumor with a poor prognosis, in which the presence of the blood-brain barrier (BBB) and skull significantly limits treatment options. To address this, a tumor-implantable optofluidic system (LED-SC), consisting of a microsized LED (microLED) and a microsyringe chip (SC), is proposed to deliver both light and prodrug nanoparticles (PNPs) directly to brain glioma. The LED-SC combines microLED and SC to enable intratumoral administration of light and PNPs for chemophotodynamic therapy. PNPs, self-assembled nanoparticles of verteporfin (VPF)-doxorubicin (DOX) prodrug, are cleaved by the enzyme cathepsin B, releasing active drugs specifically within tumor cells. In vitro studies show that PNPs are taken up by glioma cells and exhibit enhanced cytotoxicity under light irradiation. The PNPs-loaded LED-SC can be implanted into glioma, wherein PNPs are slowly diffused through the tumor, bypassing the BBB, and it also ensures effective light delivery in glioma beneath the skull, boosting chemo-photodynamic therapy. In glioma mouse models, PNP-loaded LED-SC implantation showed a 3.9-fold improvement in PNP delivery efficiency over intravenous administration, leading to better drug distribution and therapeutic results. The PNPs-loaded LED-SC offers a promising and minimally invasive solution for glioma treatment, overcoming the barriers of the BBB and skull while reducing systemic toxicity.
Collapse
Affiliation(s)
- Jagyeong Goo
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jun Seo Lee
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Junwon Park
- Department of Neurosurgery and Brain Research Institute, College of Medicine, Yonsei University, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Seong Ik Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Jeongrae Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Wan Su Yun
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Nayeon Shim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Yujeong Moon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Sunejeong Song
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Jinseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Hanhee Cho
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Ju Yeon Jeong
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Ju Seung Lee
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Sangheon Han
- Department of Neurosurgery and Brain Research Institute, College of Medicine, Yonsei University, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hyeon-Ju Lee
- Department of Neurosurgery and Brain Research Institute, College of Medicine, Yonsei University, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Won Seok Chang
- Department of Neurosurgery and Brain Research Institute, College of Medicine, Yonsei University, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Tae-Il Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
- Gradutate Program in Innovative Biomaterials Convergence, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| |
Collapse
|
7
|
Song LL, Tang YP, Qu YQ, Yun YX, Zhang RL, Wang CR, Wong VKW, Wang HM, Liu MH, Qu LQ, Wu JH, Lo HH, Law BYK. Exosomal delivery of rapamycin modulates blood-brain barrier penetration and VEGF axis in glioblastoma. J Control Release 2025; 381:113605. [PMID: 40058500 DOI: 10.1016/j.jconrel.2025.113605] [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: 06/21/2024] [Revised: 01/04/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Exosomes (Exos), nanosized membranous vesicles (30-160 nm), have been validated as an effective drug delivery system capable of traversing biological barriers. Mesenchymal stem cells (MSCs), due to their near-limitless self-renewal capabilities, provide a plentiful source of exosomes for clinical applications. In this study, we utilized an exosome-encapsulated rapamycin (Exo-Rapa) delivery strategy, which permits the use of smaller drug dosages to achieve effects typically seen with higher dosages, thus enhancing drug efficacy. Moreover, Exos can transport pharmaceuticals across the blood-brain barrier (BBB) to the brain, and further penetrate GL261 cells to exert their effects. Within the tumor microenvironment, Exo-Rapa is released more rapidly and efficiently at the tumor site. The acidic conditions in tumors accelerate the release of Exo-Rapa, a characteristic that may make it a promising targeted therapeutic in future cancer research. Additionally, a series of in vivo experiments have further demonstrated the permeability of Exo-Rapa across the BBB, enabling it to accumulate at tumor sites; it also ameliorates inflammatory responses in Glioblastoma multiforme (GBM) mouse models and enhances anti-tumor activity through the regulation of angiogenesis via the VEGF/VEGFRs axis. Our results indicate that MSC-derived exosomes are a potent therapeutic carrier for GBM, offering an effective strategy for enhancing drug delivery across the BBB and providing a scientific foundation for the use of exosomes in the treatment of GBM and other diseases.
Collapse
Affiliation(s)
- Lin Lin Song
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yong Pei Tang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yuan Qing Qu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yun Xiao Yun
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Rui Long Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Cai Ren Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Vincent Kam Wai Wong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, China
| | - Hui Miao Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Meng Han Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Li Qun Qu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jian Hui Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Hang Hong Lo
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Betty Yuen Kwan Law
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, China.
| |
Collapse
|
8
|
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
|
9
|
Jing L, Xiao W, Hu Z, Liu X, Yuan M. A Systematic Review of Nanoparticle-Mediated Ferroptosis in Glioma Therapy. Int J Nanomedicine 2025; 20:5779-5797. [PMID: 40351706 PMCID: PMC12065465 DOI: 10.2147/ijn.s523008] [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: 02/15/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Glioma, a highly malignant central nervous system tumor, exhibits aggressive invasiveness, extensive infiltration, and poor prognosis. Conventional treatments such as surgery, radiotherapy, and chemotherapy are hindered by limitations including the inability to overcome the blood-brain barrier (BBB), drug resistance, and high recurrence rates. Ferroptosis induced by nanoparticle-based systems offers an innovative strategy for glioma therapy by efficiently traversing the BBB, precisely delivering ferroptosis inducers, enhancing tumor accumulation, and enabling stimuli-responsive drug release. These features collectively improve the induction efficiency of ferroptosis in glioma cells. Various nanoplatforms, including inorganic nanoparticles, biomimetic carriers, and polymer-based systems, have demonstrated potential in crossing the BBB, inducing ferroptosis, and suppressing glioma progression. These systems enhance reactive oxygen species generation, deplete glutathione, and disrupt tumor microenvironment defense mechanisms, achieving synergistic therapeutic effects. The integration of ferroptosis with nanotechnology is emerging as a promising, non-invasive strategy for the treatment of gliomas, offering substantial therapeutic potential.
Collapse
Affiliation(s)
- Lin Jing
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Wenguang Xiao
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Zhouxing Hu
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Xu Liu
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| | - Mingqing Yuan
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, 530004, People’s Republic of China
| |
Collapse
|
10
|
Han M, He W, Zhu W, Guo L. The role of protein lactylation in brain health and disease: current advances and future directions. Cell Death Discov 2025; 11:213. [PMID: 40307243 PMCID: PMC12043837 DOI: 10.1038/s41420-025-02408-w] [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: 11/06/2024] [Revised: 02/19/2025] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Lactate, the end product of glycolysis, plays a crucial role in cellular signaling and metabolism. The discovery of lactylation, a novel post-translational modification, has uncovered the role of lactate in regulating diseases, especially in the brain. Lactylation connects genetic encoding with protein function, thereby influencing key biological processes. Increasing evidence supports lactate-mediated lactylation as a critical modulator in neurological disorders. This review offers an overview of lactate metabolism and lactylation, highlighting recent advances in understanding the regulatory enzymes of lactylation and their role in the central nervous system. We investigate the impact of lactylation on brain dysfunctions, including neurodegenerative diseases, cerebrovascular disorders, neuroinflammation, brain tumors, and psychiatric conditions. Moreover, we highlight the therapeutic potential of targeting lactylation in treating brain disorders and outline key research gaps and future directions needed to advance this promising field.
Collapse
Affiliation(s)
- Mingrui Han
- Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Queen Mary school, medical department, Nanchang University, Nanchang, Jiangxi, China
| | - Wenfeng He
- Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Wengen Zhu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Linjuan Guo
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China.
| |
Collapse
|
11
|
Din SU, Sufiyan S, Shah AA, Fatima H, Salam H, Naeem S, Laghari AA, Kayani HA, Enam SA, Mughal N. HMGA1 as a prognostic biomarker for gliomas: expression patterns, survival correlations, and clinical insights from a Pakistani cohort. J Neurooncol 2025:10.1007/s11060-025-05031-y. [PMID: 40293672 DOI: 10.1007/s11060-025-05031-y] [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: 02/03/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE High-Mobility Group A1 (HMGA1) is a chromatin-associated protein involved in regulating key cellular processes, including DNA transcription, replication, recombination, and repair. It is highly expressed during embryogenesis and reactivated in various cancers, where it contributes to tumor progression and metastasis. We investigated the prognostic significance of HMGA1 gene expression in gliomas by comparing its expression in normal brain tissue and different glioma grades. METHODS Real-time quantitative PCR (qPCR) was performed on 75 glioma samples obtained from Aga Khan University Hospital (Pakistan), along with 10 Normal Adjacent Tissue (NAT) samples. The correlation between HMGA1 expression and prognosis was evaluated using Kaplan-Meier (KM) plotter in glioma patients. Statistical analyses were conducted using the R platform and further validated through the online database Chinese Glioma Genome Atlas (CGGA) using online tools. RESULTS HMGA1 expression was significantly upregulated in gliomas compared to NAT (p < 0.001) and increased with tumor grade (p = 0.015). High HMGA1 expression correlated with Ki-67 levels and was associated with worse survival (p = 0.0014). Patients with elevated HMGA1 had a 3.5-fold higher mortality risk (95% CI: 1.5-7.9, p = 0.003). ROC analysis yielded an AUC of 0.752, indicating its potential prognostic value. CONCLUSION HMGA1 overexpression is associated with poor prognosis in gliomas, suggesting its potential as a prognostic marker. However, further validation is needed to confirm its clinical utility.
Collapse
Affiliation(s)
- Siraj Ud Din
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
- Department of Biosciences, The Shaheed Zulfikar Ali Bhutto Institute of Science and Technology, Karachi, Pakistan
| | - Sufiyan Sufiyan
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
| | - Asif Ali Shah
- Department of Biological & Biomedical Science, Aga Khan University Hospital, Karachi, Pakistan
| | - Hania Fatima
- Medical College, Aga Khan University, Karachi, Pakistan
| | - Hira Salam
- Department of Oral Pathology, Dr. Ishrat-ul-Ibad Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi, Pakistan
| | - Sana Naeem
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan
| | - Altaf Ali Laghari
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
| | - Hammad Afzal Kayani
- Department of Biosciences, The Shaheed Zulfikar Ali Bhutto Institute of Science and Technology, Karachi, Pakistan
| | - Syed Ather Enam
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan.
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan.
- Centre for Regenrative Medicine and Stem Cell Research, Aga Khan University, Karachi, Pakistan.
| | - Nouman Mughal
- Department of Biological & Biomedical Science, Aga Khan University Hospital, Karachi, Pakistan.
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan.
| |
Collapse
|
12
|
Saha S, Zhang Y, Gibert MK, Dube C, Hanif F, Mulcahy E, Bednarek S, Marcinkiewicz P, Wang X, Kwak G, Hudson K, Sun Y, Dinda M, Saha T, Guessous F, Cruickshanks N, Colon RR, Dell'Olio LG, Anbu R, Kefas B, Kumar P, Klibanov AL, Schiff D, Suk JS, Hanes J, Mata J, Hafner M, Abounader R. Discovery and therapeutic exploitation of Master Regulatory miRNAs in Glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646663. [PMID: 40236125 PMCID: PMC11996502 DOI: 10.1101/2025.04.01.646663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Glioblastoma is a fatal primary malignant brain tumor. Despite therapies involving surgical resection, chemotherapy, and radiation therapy, the average survival for glioblastoma patients remains at approximately 15 months. MicroRNAs (miRNAs) are short noncoding RNA molecules that regulate the expression of the majority of human genes. Numerous genes are concurrently deregulated in glioblastoma. Consequently, molecular monotherapies have failed to achieve improvements in clinical outcomes. Several lines of evidence suggest that simultaneous targeting of several deregulated molecules is required to achieve better therapies. However, the simultaneous targeting of several deregulated oncogenic drivers is severely limited by the fact that the drugs needed to target many deregulated molecules do not currently exist, and because combining several drugs in a clinical setting leads to an exponential increase in toxicity. We hypothesized that we can develop and use miRNA to simultaneously inhibit multiple deregulated genes for more efficacious glioblastoma therapies. The goal of this study was therefore to identify master regulatory microRNAs (miRNAs) and use them to simultaneously target multiple deregulated molecules for GBM therapy. We defined master regulatory miRNAs as those that target several deregulated genes in glioblastoma. To find master regulatory miRNAs, we first used PAR-CLIP screenings to identify all targets of all miRNAs in glioblastoma cells. We then analyzed TCGA tumor data to determine which of these targets are deregulated in human tumors. We developed and used an algorithm to rank these targets for significance in glioblastoma malignancy based on their magnitude of deregulation, frequency of deregulation, and correlation with patient survival. We then ranked the miRNAs for their capacity of targeting multiple glioblastoma-deregulated genes and therefore the potential to exhibit strong anti-tumor effects when delivered as therapy. Using this strategy, we selected two tumor suppressor master regulatory miRNAs, miR-340, miR-382 and an oncogenic master regulatory miRNA, miR-17. We validated the target genes of the miRNAs and showed that they form part of important glioblastoma regulatory pathways. We then showed that the miRNAs (miR-340 and miR-582) or the miR-17 inhibitor have strong inhibitory effects on glioblastoma cell growth, survival, invasion, stemness and in vivo tumor growth. Ultimately, we developed and successfully tested a new therapeutic approach to delivery miR-340 using MRI guided focused ultrasound and microbubbles (FUS-MB) and special brain penetrating nanoparticles (BPN). This approach resulted in a substantial reduction in tumor volume and prolongation of the survival of glioblastoma-bearing mice and can be translated into clinical trials. We therefore developed and successfully tested a novel strategy to discover and deliver miRNAs for glioblastoma and cancer therapy.
Collapse
|
13
|
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
|
14
|
Mandal M, Banerjee I, Mandal M. Effective approaches in conquering chemoresistance of glioblastoma: potential for nanoformulations. Drug Deliv Transl Res 2025:10.1007/s13346-025-01859-z. [PMID: 40259195 DOI: 10.1007/s13346-025-01859-z] [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] [Accepted: 04/10/2025] [Indexed: 04/23/2025]
Abstract
Glioblastoma Multiforme is an aggressive and complex cancer affecting mostly elderly patients above the age of 60 years. Originally classified as the fourth stage of glioma, it has an abysmal prognosis along with limited therapeutic options. Surgical removal of tumors, radiotherapy, and chemotherapy are prevalent treatment strategies with numerous therapeutic obstacles, including undefined boundary of tumor mass leaving traces even after excision, chances of secondary cancer formation, and presence of blood-brain barrier. These blood-brain and blood-brain tumor barriers actively restrict the permeability of many molecules from blood circulation to enter the central nervous system. Therefore, many conventional antineoplastic drugs fail to reach the tumor periphery except temozolomide. Meanwhile, active stem cells in the tumor microenvironment, genetic mutation inducing tumor growth, and epigenetic pattern alteration make this cancer chemoresistant. Our review delineates the recent approaches to resensitize the existing clinical drugs through specifically designed nanoformulations. Nanoparticles with modified physiological characteristics and modified through technological parameters can reduce the tumor's stemness, which increases tumor cells' apoptosis rate. Moreover, these nanoparticles can efficiently traverse the blood-brain barrier and escape from endosomal degradation with minimum toxicological impact. Overall, this review discusses the cancer chemoresistance phenomena and related pathways and highlights the potential of nanoformulation in reversing chemoresistance. Also, the existing limitations of this unique approach and suggestions are discussed at the end of the article, which may facilitate the identification of new directions for advancement of the nanoparticle-mediated reversal of chemoresistance.
Collapse
Affiliation(s)
- Madhurima Mandal
- School of Medical Science & Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India
| | - Indranil Banerjee
- Department of Pharmaceutical Technology, JIS University, 81, Nilgunj Road, Agarpara, Kolkata, West Bengal, 700109, India.
| | - Mahitosh Mandal
- School of Medical Science & Technology, Indian Institute of Technology, Kharagpur, West Bengal, 721302, India.
| |
Collapse
|
15
|
Batista C, Cruz JVR, Siqueira M, Pesquero JB, Stipursky J, Mendes FDA. Kinin B 1 Receptor Agonist Enhances Blood-Brain Barrier Permeability in Healthy and Glioblastoma Environments. Pharmaceuticals (Basel) 2025; 18:591. [PMID: 40284027 PMCID: PMC12030169 DOI: 10.3390/ph18040591] [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: 02/27/2025] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The low permeability of the blood-brain barrier (BBB) represents a significant challenge to effective systemic chemotherapy for primary and metastatic brain cancers. Kinin receptors play a crucial role in modulating BBB permeability, and their agonist analogs have been explored in preclinical animal models to enhance drug delivery to the brain. In this study, we investigated whether des-Arg9-bradykinin (DBK), a physiological agonist of kinin B1 receptor (B1R), acts as a brain drug delivery adjuvant by promoting the transient opening of the BBB. Methods: Human brain microvascular endothelial cells (HBMECs) were treated with DBK in the culture medium and in conditioned media from glioblastoma cell lines, namely T98G (CMT98G) and U87MG (CMU87). Immunofluorescence, RT-qPCR, in-cell Western assay, and proximity ligation assay (PLA) were performed to analyze BBB components, kinin receptors and TLR4, a receptor associated with the kinin pathway and inflammation. The effect of DBK on enhancing paracellular molecule transport was evaluated using Evans blue dye (EB) quantification in a cell culture insert assay and in an in vivo model, where mice with and without brain tumors were treated with DBK. To assess the functional impact of the transient BBB opening induced by DBK, the chemotherapeutic drug doxorubicin (DOX) was administered. Results: Treatment with DBK facilitates the presence of EB in the brain parenchyma by transiently disrupting the BBB, as further evidenced by the increased paracellular passage of the dye in an in vitro assay. B1R activation by DBK induces transient BBB opening lasting less than 48 h, enhancing the bioavailability of the DOX within the brain parenchyma and glioma tumor mass. The interaction between B1R and TLR4 is disrupted by the secreted factors released by glioblastoma cells, as conditioned media from T98G and U87 reduce TLR4 staining in endothelial cells without affecting B1R expression. Conclusions: These results further support the potential of B1R activation as a strategy to enhance targeted drug delivery to the brain.
Collapse
Affiliation(s)
- Carolina Batista
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (C.B.); (J.V.R.C.); (M.S.); (J.S.)
| | - João Victor Roza Cruz
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (C.B.); (J.V.R.C.); (M.S.); (J.S.)
| | - Michele Siqueira
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (C.B.); (J.V.R.C.); (M.S.); (J.S.)
| | - João Bosco Pesquero
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo 04039-032, SP, Brazil;
| | - Joice Stipursky
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (C.B.); (J.V.R.C.); (M.S.); (J.S.)
| | - Fabio de Almeida Mendes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (C.B.); (J.V.R.C.); (M.S.); (J.S.)
| |
Collapse
|
16
|
Liu J, Wang Z. The landscape of FGFR-TACC fusion in adult glioblastoma: From bench to bedside. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2025; 795:108536. [PMID: 40246063 DOI: 10.1016/j.mrrev.2025.108536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Glioblastoma (GBM) is a lethal central nervous system tumor, characterized by extensive genomic alterations and high intra-tumoral heterogeneity. Gene fusions, derived from chromosomal translocations, deletions, and inversions, were increasingly recognized as key carcinogenic events, with the highest frequency of FGFR-TACC fusion in glioblastoma. As reported, FGFR3-TACC3 fusion mostly coexists with wild-type IDH status, and associates with better prognosis. Mechanistically, FGFR3-TACC3 fusions can constitutively activate non-canonical FGFR downstream pathways, induce aneuploidy, and participate in mitochondrial metabolism, thereby promoting cell proliferation and tumorigenesis. These functions, whether based on FGFR3 phosphorylation or not, are predominantly attributed to the specific domain of TACC3 that involved in regulating the localization and activation of fusion products. Several preclinical studies and clinical trials are being performed to evaluate the efficacy and safety of the FGFR-TACC fusion as a personalised therapeutic target, including the treatments with tyrosine kinase inhibitors, metabolic inhibitors, HSP90 inhibitors, coiled-coil peptide-mimetics, and targeted protein degraders. A subset of populations with FGFR-TACC-positive glioblastoma, after refined molecular screening strategies, may benefit from targeted therapies. Despite major progress in biotechnology, our understanding on the role of fusion events in glioblastoma represented by the FGFR-TACC is still in its infancy. Here, we highlight recent progress on FGFR-TACC fusion in human glioblastoma, emphasizing their molecular mechanisms and potential clinical value.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiotherapy, Tianjin First Central Hospital, Nankai University, Tianjin 300384, China
| | - Zheng Wang
- Department of Radiotherapy, Tianjin First Central Hospital, Nankai University, Tianjin 300384, China.
| |
Collapse
|
17
|
Tang J, Amin MA, Campian JL. Glioblastoma Stem Cells at the Nexus of Tumor Heterogeneity, Immune Evasion, and Therapeutic Resistance. Cells 2025; 14:562. [PMID: 40277888 PMCID: PMC12025403 DOI: 10.3390/cells14080562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/26/2025] Open
Abstract
Glioblastoma (GBM) is an exceedingly aggressive primary brain tumor defined by rapid growth, extensive infiltration, and resistance to standard therapies. A central factor driving these malignancies is the subpopulation of glioblastoma stem cells (GSCs), which possess self-renewal capacity, multipotency, and the ability to regenerate tumor heterogeneity. GSCs contribute to key hallmarks of GBM pathobiology, including relentless progression, resistance to chemotherapy and radiotherapy, and inevitable recurrence. GSCs exhibit distinct molecular signatures, enhanced DNA repair, and metabolic adaptations that protect them against conventional treatments. Moreover, they reside within specialized niches-such as perivascular or hypoxic microenvironments-that sustain stemness, promote immunosuppression, and facilitate angiogenesis. Recent discoveries highlight signaling pathways like Notch, Wnt/β-catenin, Hedgehog, STAT3-PARN, and factors such as TFPI2 and HML-2 as critical regulators of GSC maintenance, plasticity, and immune evasion. These findings underscore the complexity of GSC biology and their pivotal role in driving GBM heterogeneity and therapeutic failure. Emerging therapeutic strategies aim to target GSCs through multiple avenues, including surface markers, immunotherapeutics (e.g., CAR T cells), metabolic vulnerabilities, and combination regimens. Advances in patient-derived organoids, single-cell omics, and 3D co-culture models enable more accurate representation of the tumor ecosystem and personalized therapeutic approaches. Ultimately, improved understanding of GSC-specific targets and the tumor microenvironment promises more effective interventions, paving the way toward better clinical outcomes for GBM patients.
Collapse
Affiliation(s)
- Justin Tang
- Department of Biomedical Science, University of Guelph, Guelph, ON N1G 2W1, Canada
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (M.A.A.); (J.L.C.)
| | - Md Al Amin
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (M.A.A.); (J.L.C.)
| | - Jian L. Campian
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (M.A.A.); (J.L.C.)
| |
Collapse
|
18
|
Yoshino Y, Yoshino F, Aoki I, Mori Y, Suzuki G, Tsuji S, Amano T, Shiino A, Chano T, Furusho Y, Murakami T, Yamazaki H, Yamada K. 2-Nitroimidazole-Functionalized Superparamagnetic Iron Oxide Nanoparticles Detect Hypoxic Regions of Glioblastomas on MRI and Improve Radiotherapy Efficacy. ACS NANO 2025; 19:12762-12776. [PMID: 40139197 PMCID: PMC11984306 DOI: 10.1021/acsnano.4c06753] [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: 05/21/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
The presence of hypoxic regions in tumors is associated with malignancy and is an important target for the high-precision diagnosis and treatment of tumors. Radioresistant hypoxic regions can be precisely identified and treated without the use of high doses of radiation if hypoxic region-specific contrast agents have a therapeutic effect. In this study, we synthesized a therapeutic-diagnostic complex agent (SPION-PG-NI) by combining polyglycerol-functionalized superparamagnetic iron oxide nanoparticles (SPION-PG, core diameter of 8.8 ± 1.9 nm) as an MRI contrast agent and 2-nitroimidazole (NI, a pimonidazole derivative) as a hypoxia-targeted ligand to visually evaluate hypoxic regions using MRI and improve radiotherapy efficacy at those sites. SPION-PG-NI showed a concentration-dependent contrast effect and had significantly higher accumulation in subcutaneous glioblastomas than the control agent, SPION-PG, 24 h after administration. Immunohistological evaluations showed that the SPION-PG-NI-accumulated regions corresponded well to hypoxic regions. SPION-PG-NI showed neither migration into the brain parenchyma nor neurotoxicity. Both SPION-PG and SPION-PG-NI decrease reactive oxygen species (ROS); however, they improve radiotherapy efficacy in hypoxic glioblastoma cells due to cytotoxicity. This effect of SPION-PG-NI was significantly higher than that of SPION-PG (p < 0.01). After 12 Gy irradiation, the mean normalized glioblastoma tumor volume on day 38 in the SPION-PG-NI group (288%) was significantly lower than that in the control group (882%) (p < 0.05). Collectively, these findings suggest the potential of SPION-PG-NI as a useful and safe tumor theranostic nanodevice for hypoxic imaging and improving radiotherapy efficacy.
Collapse
Affiliation(s)
- Yuki Yoshino
- Department
of Radiology, Kyoto Prefectural University
of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Kansai
BNCT Medical Center, Educational Foundation
of Osaka Medical and Pharmaceutical University, 2-7 Daigakucho, Takatsuki Osaka 569-8686, Japan
| | - Fumi Yoshino
- Department
of Obstetrics and Gynecology, Shiga University
of Medical Science, Seta, Otsu 520-2192, Japan
- Mariko
Clinic, 13-5 Noji, Kusatsu 525-0059, Japan
| | - Ichio Aoki
- Institute
for Quantum Medical Science, National Institutes
for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage 263-8555 Chiba, Japan
| | - Yasuyuki Mori
- Department
of Chemistry, Shiga University of Medical
Science, Otsu 520-2192, Japan
| | - Gen Suzuki
- Department
of Radiology, Kyoto Prefectural University
of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shunichiro Tsuji
- Department
of Obstetrics and Gynecology, Shiga University
of Medical Science, Seta, Otsu 520-2192, Japan
| | - Tsukuru Amano
- Department
of Obstetrics and Gynecology, Shiga University
of Medical Science, Seta, Otsu 520-2192, Japan
| | - Akihiko Shiino
- Department
of Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Tokuhiro Chano
- Department
of Clinical Laboratory Medicine, Shiga University
of Medical Science, Seta, Otsu 520-2192, Japan
| | - Yoshio Furusho
- Department
of Chemistry, Shiga University of Medical
Science, Otsu 520-2192, Japan
| | - Takashi Murakami
- Department
of Obstetrics and Gynecology, Shiga University
of Medical Science, Seta, Otsu 520-2192, Japan
| | - Hideya Yamazaki
- Department
of Radiology, Kyoto Prefectural University
of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kei Yamada
- Department
of Radiology, Kyoto Prefectural University
of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
19
|
Jurgaitis J, Mickeviciute K, Jablonskiene V. Overcoming the Blood-Brain Barrier: Focused Ultrasound in Glioblastoma Treatment. Cureus 2025; 17:e82869. [PMID: 40416199 PMCID: PMC12103647 DOI: 10.7759/cureus.82869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/27/2025] Open
Abstract
Despite recent advancements in neuro-oncology, glioblastoma remains one of the cancers with the poorest prognosis. Tumor recurrence and progression are driven by tumor stem cells and a high mutational burden. New potential treatment modalities are actively being explored. One promising approach is focused ultrasound (FUS), which involves converging ultrasound waves on a specific region of interest while avoiding damage to surrounding tissue. This mini-review highlights the current understanding of glioblastoma treatment resistance and positions FUS as a promising therapeutic option. We discuss existing clinical research utilizing FUS in four key treatment applications: (1) blood-brain barrier disruption, (2) histotripsy, (3) thermal ablation, and (4) sonodynamic therapy.
Collapse
Affiliation(s)
- Jonas Jurgaitis
- Department of Neurosurgery and Neurology, Faculty of Medicine, Vilnius University, Vilnius, LTU
| | - Karina Mickeviciute
- Department of Neuroradiology, Faculty of Medicine, Vilnius University, Vilnius, LTU
| | - Valerija Jablonskiene
- Department of Physiology, Biochemistry, Microbiology, and Laboratory Medicine, Faculty of Medicine, Vilnius University, Vilnius, LTU
| |
Collapse
|
20
|
Ding Y, Chen R, Zhou J, Bao Y, Meng N, Zheng X, Yang S, Lu J, Jiang Z, Liu Y, Xie C, Lu L, Lu W. All-stage targeted nanodiscs for glioma treatment by inducing cuproptosis and apoptosis of cancer cells and cancer stem cells. Asian J Pharm Sci 2025; 20:101010. [PMID: 40182135 PMCID: PMC11964743 DOI: 10.1016/j.ajps.2024.101010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/21/2024] [Accepted: 11/05/2024] [Indexed: 04/05/2025] Open
Abstract
There remain several intractable challenges for chemotherapy in glioma treatment, including the blood-brain barrier (BBB), blood-brain tumor barrier (BBTB), and tumor heterogeneity caused by cancer stem cells (CSCs), which are resistant to conventional chemotherapy. Here, we established a nano strategy to kill glioma cells and CSCs, combining carfilzomib and bis(diethyldithiocarbamate)copper. The synergistic drug combination disturbed cell protein metabolism at different stages and induced apoptosis and cuproptosis. The Y-shaped targeting ligand pHA-VAP-modified nanodiscs were designed to help the chemotherapeutic agents cross the BBB/BBTB and finally accumulate in tumor site. This all-stage targeting and all-stage treatment nanomedicine significantly prolonged the survival in glioma-bearing mice and might inspire the rational design of advanced drug delivery platforms.
Collapse
Affiliation(s)
- Yuan Ding
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Ruohan Chen
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jianfen Zhou
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yanning Bao
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Nana Meng
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Xudong Zheng
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Shengmin Yang
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jiasheng Lu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Zhixuan Jiang
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yu Liu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Cao Xie
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Weiyue Lu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China
| |
Collapse
|
21
|
Luo Q, Yang J, Yang M, Wang Y, Liu Y, Liu J, Kalvakolanu DV, Cong X, Zhang J, Zhang L, Guo B, Duo Y. Utilization of nanotechnology to surmount the blood-brain barrier in disorders of the central nervous system. Mater Today Bio 2025; 31:101457. [PMID: 39896289 PMCID: PMC11786670 DOI: 10.1016/j.mtbio.2025.101457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/27/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Central nervous system (CNS) diseases are a major cause of disability and death worldwide. Due to the blood-brain barrier (BBB), drug delivery for CNS diseases is extremely challenging. Nano-delivery systems can overcome the limitations of BBB to deliver drugs to the CNS, improve the ability of drugs to target the brain and provide potential therapeutic methods for CNS diseases. At the same time, the choice of different drug delivery methods (bypassing BBB or crossing BBB) can further optimize the therapeutic effect of the nano-drug delivery system. This article reviews the different methods of nano-delivery systems to overcome the way BBB enters the brain. Different kinds of nanoparticles to overcome BBB were discussed in depth.
Collapse
Affiliation(s)
- Qian Luo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Jiaying Yang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Mei Yang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Yingtong Wang
- The Undergraduate Center of Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Yiran Liu
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Jixuan Liu
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Dhan V. Kalvakolanu
- Greenebaum NCI Comprehensive Cancer Center, Department of Microbiology and Immunology University of Maryland School Medicine, Baltimore, MD, USA
| | - Xianling Cong
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Jinnan Zhang
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Ling Zhang
- Key Laboratory of Pathobiology, Ministry of Education, And Department of Biomedical Science, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Baofeng Guo
- Department of Plastic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Yanhong Duo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
22
|
Fu M, Xue B, Miao X, Gao Z. Overcoming immunotherapy resistance in glioblastoma: challenges and emerging strategies. Front Pharmacol 2025; 16:1584688. [PMID: 40223940 PMCID: PMC11987931 DOI: 10.3389/fphar.2025.1584688] [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: 02/27/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults, characterized by rapid proliferation, extensive infiltration, and significant intratumoral heterogeneity. Despite advancements in conventional treatments, including surgery, radiotherapy, and chemotherapy, the prognosis for GBM patients remains poor, with a median survival of approximately 15 months. Immunotherapy has emerged as a promising alternative; however, the unique biological and immunological features, including its immunosuppressive tumor microenvironment (TME) and low mutational burden, render it resistant to many immunotherapeutic strategies. This review explores the key challenges in GBM immunotherapy, focusing on immune evasion mechanisms, the blood-brain barrier (BBB), and the TME. Immune checkpoint inhibitors and CAR-T cells have shown promise in preclinical models but have limited clinical success due to antigen heterogeneity, immune cell exhaustion, and impaired trafficking across the BBB. Emerging strategies, including dual-targeting CAR-T cells, engineered immune cells secreting therapeutic molecules, and advanced delivery systems to overcome the BBB, show potential for enhancing treatment efficacy. Addressing these challenges is crucial for improving GBM immunotherapy outcomes.
Collapse
Affiliation(s)
- Maowu Fu
- Department of Neurosurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Bing Xue
- Department of Neurosurgery, Jinan Third People’s Hospital, Jinan, Shandong, China
| | - Xiuming Miao
- Department of Pathology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zong Gao
- Department of Neurosurgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
23
|
Chen X, Zhong X, Zhang F, Zhou X, Yue X, Li X. Molecular mechanisms and therapeutic targets in glioblastoma multiforme: network and single-cell analyses. Sci Rep 2025; 15:10558. [PMID: 40148380 PMCID: PMC11950307 DOI: 10.1038/s41598-025-92867-z] [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: 09/01/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain tumor associated with poor survival outcomes and is driven by a complex tumor microenvironment (TME) that promotes tumor progression and treatment resistance. To explore the role of the TME in GBM, we analyzed glioma-related microarray and single-cell RNA sequencing (scRNA-seq) datasets from the Gene Expression Omnibus (GEO). Functional enrichment and weighted gene coexpression network analyses revealed distinct immune profiles, metabolic alterations, and differences in chemotherapeutic drug sensitivity between the high-risk and low-risk patient groups. scRNA-seq data processed with the 'Seurat' package were used to identify differentially expressed genes in pericytes, endothelial cells, and glioma cells, particularly those involved in extracellular matrix (ECM) remodeling. A 17-gene prognostic signature developed through Cox regression and LASSO analyses revealed that key genes (COL1A1, COL4A1, and VIM) were significantly associated with survival outcomes in GBM patients. Drug sensitivity analyses using data from the Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Therapeutics Response Portal (CTRP) identified potential targeted therapies for GBM, including SB-505,124, staurosporine, and AZD8186. This integrative study underscores the critical roles of the ECM and synaptic remodeling in GBM and suggests novel therapeutic targets to improve personalized treatment strategies for GBM patients.
Collapse
Affiliation(s)
- Xiangyu Chen
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400016, China
| | - Xiao Zhong
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400016, China
| | - Feifei Zhang
- Department of Blood Transfusion, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610021, Sichuan, China
| | - Xiaomei Zhou
- Sichuan Provincial Chengdu Second People's Hospital, Chengdu, 610021, Sichuan, China
| | - Xiaofeng Yue
- Department of Urology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China.
| | - Xueru Li
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
24
|
Beylerli O, Gareev I, Musaev E, Roumiantsev S, Chekhonin V, Ahmad A, Chao Y, Yang G. New approaches to targeted drug therapy of intracranial tumors. Cell Death Discov 2025; 11:111. [PMID: 40113789 PMCID: PMC11926108 DOI: 10.1038/s41420-025-02358-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
Intracranial tumors encompass a heterogeneous group of neoplasms, including gliomas, meningiomas, pituitary adenomas, schwannomas, craniopharyngiomas, ependymomas, medulloblastomas, and primary central nervous system lymphomas. These tumors present significant challenges due to their diverse molecular characteristics, critical locations, and the unique obstacles posed by the blood-brain barrier (BBB) and blood-tumor barrier (BTB), which limit the efficacy of systemic therapies. Recent advances in molecular biology and genomics have enabled the identification of specific molecular pathways and targets, paving the way for innovative precision therapies. This review examines the current state of targeted therapies for intracranial tumors, including receptor tyrosine kinase (RTK) inhibitors, PI3K/AKT/mTOR inhibitors, RAF/MEK/ERK pathway inhibitors, IDH mutation inhibitors, immune checkpoint inhibitors, and CAR-T cell therapies. Emphasis is placed on the role of the BBB and BTB in modulating drug delivery and therapeutic outcomes. Strategies to overcome these barriers, such as focused ultrasound, nanoparticle-based delivery systems, and convection-enhanced delivery, are also explored. Furthermore, the manuscript reviews clinical trial data, highlighting successes and limitations across different tumor types. It delves into emerging therapeutic approaches, including combination of regimens and personalized treatments based on molecular profiling. By synthesizing the latest research, this article aims to provide a comprehensive understanding of the advancements and ongoing challenges in the targeted treatment of intracranial tumors. The findings underscore the necessity for innovative delivery systems and more extensive clinical trials to optimize therapeutic strategies. This review aspires to inform future research and clinical practices, aiming to improve patient outcomes and quality of life in the management of these complex and life-threatening conditions.
Collapse
Affiliation(s)
- Ozal Beylerli
- Central Research Laboratory, Bashkir State Medical University, Ufa, Republic of Bashkortostan, Russian Federation.
| | - Ilgiz Gareev
- Central Research Laboratory, Bashkir State Medical University, Ufa, Republic of Bashkortostan, Russian Federation
| | - Elmar Musaev
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Sergey Roumiantsev
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
| | - Vladimir Chekhonin
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Endocrinology Research Center, Moscow, Russian Federation
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Yuan Chao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Heilongjiang Province Neuroscience Institute, Harbin, China
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Heilongjiang Province Neuroscience Institute, Harbin, China.
| |
Collapse
|
25
|
Paolani G, Minosse S, Strolin S, Santoro M, Pucci N, Di Giuliano F, Garaci F, Oddo L, Toumia Y, Guida E, Riccitelli F, Perilli G, Vitaliti A, Bedini A, Dolci S, Paradossi G, Domenici F, Da Ros V, Strigari L. Intra-Arterial Super-Selective Delivery of Yttrium-90 for the Treatment of Recurrent Glioblastoma: In Silico Proof of Concept with Feasibility and Safety Analysis. Pharmaceutics 2025; 17:345. [PMID: 40143008 PMCID: PMC11945926 DOI: 10.3390/pharmaceutics17030345] [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: 01/11/2025] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Intra-arterial cerebral infusion (IACI) of radiotherapeutics is a promising treatment for glioblastoma (GBM) recurrence. We investigated the in silico feasibility and safety of Yttrium-90-Poly(vinyl alcohol)-Microbubble (90Y-PVA-MB) IACI in patients with recurrent GBM and compared the results with those of external beam radiation therapy (EBRT). Methods: Contrast-enhanced T1-weighted magnetic resonance imaging (T1W-MRI) was used to delineate the tumor volumes and CT scans were used to automatically segment the organs at risk in nine patients with recurrent GBM. Volumetric Modulated Arc Therapy (VMAT) treatment plans were generated using a clinical treatment planning system. Assuming the relative intensity of each voxel from the MR-T1W as a valid surrogate for the post-IACI 90Y-PVA-MB distribution, a specific 90Y dose voxel kernel was obtained through Monte Carlo (MC) simulations and convolved with the MRI, resulting in a 90Y-PVA-MB-based dose distribution that was then compared with the VMAT plans. Results: The physical dose distribution obtained from the simulation of 1GBq of 90Y-PVA-MBs was rescaled to ensure that 95% of the prescribed dose was delivered to 95% or 99% of the target (i.e., A95% and A99%, respectively). The calculated activities were A95% = 269.2 [63.6-2334.1] MBq and A99% = 370.6 [93.8-3315.2] MBq, while the mean doses to the target were 58.2 [58.0-60.0] Gy for VMAT, and 123.1 [106.9-153.9] Gy and 170.1 [145.9-223.8] Gy for A95% and A99%, respectively. Additionally, non-target brain tissue was spared in the 90Y-PVA-MB treatment compared to the VMAT approach, with a median [range] of mean doses of 12.5 [12.0-23.0] Gy for VMAT, and 0.6 [0.2-1.0] Gy and 0.9 [0.3-1.5] Gy for the 90Y treatments assuming A95% and A99%, respectively. Conclusions: 90Y-PVA-MB IACI using MR-T1W appears to be feasible and safe, as it enables the delivery of higher doses to tumors and lower doses to non-target volumes compared to the VMAT approach.
Collapse
Affiliation(s)
- Giulia Paolani
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (S.S.); (M.S.); (L.S.)
| | - Silvia Minosse
- U.O.C Diagnostic Imaging, Department of Integrated Care Processes, Fondazione PTV Policlinico “Tor Vergata”, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy;
| | - Silvia Strolin
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (S.S.); (M.S.); (L.S.)
| | - Miriam Santoro
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (S.S.); (M.S.); (L.S.)
| | - Noemi Pucci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy; (N.P.); (F.D.G.); (F.G.); (E.G.); (S.D.); (V.D.R.)
| | - Francesca Di Giuliano
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy; (N.P.); (F.D.G.); (F.G.); (E.G.); (S.D.); (V.D.R.)
| | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy; (N.P.); (F.D.G.); (F.G.); (E.G.); (S.D.); (V.D.R.)
| | - Letizia Oddo
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (L.O.); (Y.T.); (F.R.); (G.P.); (A.V.); (G.P.)
| | - Yosra Toumia
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (L.O.); (Y.T.); (F.R.); (G.P.); (A.V.); (G.P.)
- National Institute for Nuclear Physics (INFN), sez. Roma Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Eugenia Guida
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy; (N.P.); (F.D.G.); (F.G.); (E.G.); (S.D.); (V.D.R.)
| | - Francesco Riccitelli
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (L.O.); (Y.T.); (F.R.); (G.P.); (A.V.); (G.P.)
| | - Giulia Perilli
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (L.O.); (Y.T.); (F.R.); (G.P.); (A.V.); (G.P.)
| | - Alessandra Vitaliti
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (L.O.); (Y.T.); (F.R.); (G.P.); (A.V.); (G.P.)
| | - Angelico Bedini
- Department of Technological Innovations and Safety of Plants, Products and Anthropic Settlements (DIT), Italian National Institute for Insurance against Accidents at Work, Inail, Piazzale Giulio Pastore 6, 00144 Rome, Italy;
| | - Susanna Dolci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy; (N.P.); (F.D.G.); (F.G.); (E.G.); (S.D.); (V.D.R.)
| | - Gaio Paradossi
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (L.O.); (Y.T.); (F.R.); (G.P.); (A.V.); (G.P.)
- National Institute for Nuclear Physics (INFN), sez. Roma Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Fabio Domenici
- Department of Chemical Science and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (L.O.); (Y.T.); (F.R.); (G.P.); (A.V.); (G.P.)
- National Institute for Nuclear Physics (INFN), sez. Roma Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Valerio Da Ros
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy; (N.P.); (F.D.G.); (F.G.); (E.G.); (S.D.); (V.D.R.)
| | - Lidia Strigari
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.P.); (S.S.); (M.S.); (L.S.)
| |
Collapse
|
26
|
Zhang J, Yan X, Gao X, Hu J, Zhao H, Yang C, Xu L, Niu Y, Geng M, Wang N, Hu S. Three Decades of Photodynamic Therapy for Glioblastoma: A Comprehensive Scientometric Analysis. Photodiagnosis Photodyn Ther 2025; 53:104533. [PMID: 40054646 DOI: 10.1016/j.pdpdt.2025.104533] [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: 12/13/2024] [Revised: 02/09/2025] [Accepted: 02/26/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Photodynamic therapy (PDT) has emerged as a promising adjunctive treatment for glioblastoma (GBM), yet a comprehensive bibliometric analysis of this field is lacking. This study explores research trends, major contributors, and hotspots in PDT for GBM to provide an integrated overview of its development. METHODS Literature from 1993 to 2024 was retrieved from the Web of Science Core Collection. Bibliometric tools, including CiteSpace, analyzed publication trends, collaborations, and keyword co-occurrence to identify influential authors, institutions, and journals. RESULTS A total of 799 publications showed a growing research interest, peaking in 2022. The United States and China were leading contributors, with prominent institutions like the University of California System and Centre National de la Recherche Scientifique. Influential figures, such as Jiro Akimoto and Walter Stummer, advanced clinical applications and fluorescence-guided techniques. Early studies of PDT for GBM have focused on evaluating its efficacy and potential side effects, while recent research has transitioned toward innovative strategies like targeted drug delivery, nanotechnology, and combination therapies. However, the similarities between early and recent studies are in the search for safe and reliable photosensitizers. Keyword analysis highlighted "5-aminolevulinic acid", "in vitro", and "polyethylene glycol compounds" as key areas, while timeline analysis revealed shifts from foundational photosensitizer research to approaches addressing tumor heterogeneity and resistance. CONCLUSIONS This study provides a systematic overview of PDT research for GBM, spotlighting breakthroughs and collaborative networks. The findings emphasize the importance of innovation and clinical translation to fully realize PDT's potential in GBM therapy.
Collapse
Affiliation(s)
- Jiheng Zhang
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiuwei Yan
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xin Gao
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiahe Hu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Hongtao Zhao
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chengyun Yang
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lei Xu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yapeng Niu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Mo Geng
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Nan Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Shaoshan Hu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
27
|
Mi Y, Jiang P, Luan J, Feng L, Zhang D, Gao X. Peptide‑based therapeutic strategies for glioma: Current state and prospects. Peptides 2025; 185:171354. [PMID: 39922284 DOI: 10.1016/j.peptides.2025.171354] [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: 11/23/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Glioma is a prevalent form of primary malignant central nervous system tumor, characterized by its cellular invasiveness, rapid growth, and the presence of the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB). Current therapeutic approaches, such as chemotherapy and radiotherapy, have shown limited efficacy in achieving significant antitumor effects. Therefore, there is an urgent demand for new treatments. Therapeutic peptides represent an innovative class of pharmaceutical agents with lower immunogenicity and toxicity. They are easily modifiable via chemical means and possess deep tissue penetration capabilities which reduce side effects and drug resistance. These unique pharmacokinetic characteristics make peptides a rapidly growing class of new therapeutics that have demonstrated significant progress in glioma treatment. This review outlines the efforts and accomplishments in peptide-based therapeutic strategies for glioma. These therapeutic peptides can be classified into four types based on their anti-tumor function: tumor-homing peptides, inhibitor/antagonist peptides targeting cell surface receptors, interference peptides, and peptide vaccines. Furthermore, we briefly summarize the results from clinical trials of therapeutic peptides in glioma, which shows that peptide-based therapeutic strategies exhibit great potential as multifunctional players in glioma therapy.
Collapse
Affiliation(s)
- Yajing Mi
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Pengtao Jiang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Jing Luan
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lin Feng
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Dian Zhang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Xingchun Gao
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China.
| |
Collapse
|
28
|
Khafaga DSR, Muteeb G, Aswa DW, Aatif M, Farhan M, Allam S. Green chemistry: Modern therapies using nanocarriers for treating rare brain cancer metastasis from colon cancer. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100213. [PMID: 39826871 DOI: 10.1016/j.slasd.2025.100213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Brain metastasis (BM) from colon cancer is associated with a poor prognosis and restricted treatment alternatives, largely due to issues related to blood-brain barrier (BBB) permeability and the negative effects of standard chemotherapy. Nanotechnology improves treatment efficacy by enabling targeted and controlled drug delivery. This review article evaluates the potential of nanotechnology-based therapies for treating colon cancer BM, emphasizing their capacity to cross the BBB, diminish metastatic growth, and enhance overall survival rates. A review of multiple studies evaluated nanoparticles (NPs) as carriers for chemotherapy, focusing on parameters including particle size, surface charge, and drug-loading capacity. The study also reviewed studies that examined BBB penetration, in vitro tumor accumulation, and in vivo tumor growth inhibition. In vitro findings indicated that NPs accumulate more efficiently in BM tissue than in healthy brain tissue and show significant BBB penetration. In vivo, nanotherapy markedly inhibited tumor growth and prolonged survival relative to conventional chemotherapy or control treatments while also exhibiting reduced side effects. Recent studies demonstrated that plant extracts can effectively and safely synthesize nanomaterials, positioning them as a viable and environmentally friendly precursor for nanomaterial production. Nanotechnology-based therapies demonstrate significant potential in the treatment of colon cancer BM by minimizing systemic toxicity, enhancing therapeutic efficacy, and facilitating more targeted drug delivery. Further research is required to confirm these findings and implement them in clinical practice.
Collapse
Affiliation(s)
- Doaa S R Khafaga
- Health Sector, Faculty of Science, Galala University, New Galala City 43511, Suez, Egypt.
| | - Ghazala Muteeb
- Department of Nursing, College of Applied Medical Sciences, King Faisal University, Al-Ahsa, Saudi Arabia.
| | - Darin W Aswa
- Faculty of Medicine, Galala University, New Galala City 43511, Suez, Egypt
| | - Mohammad Aatif
- Department of Public Health, College of Applied Medical Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Mohd Farhan
- Department of Basic Sciences, Preparatory Year, King Faisal University, Al-Ahsa 31982, Saudi Arabia; Department of Chemistry, College of Science, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Salma Allam
- Faculty of Medicine, Galala University, New Galala City 43511, Suez, Egypt
| |
Collapse
|
29
|
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
|
30
|
Gai S, Yan Q, Li S, Zhong X, Qin Y, Jiang M. Lactoferrin Nanoparticle-Vanadium Complex: A Promising High-Efficiency Agent against Glioblastoma by Triggering Autophagy and Ferroptosis. J Med Chem 2025; 68:4650-4662. [PMID: 39945608 DOI: 10.1021/acs.jmedchem.4c02696] [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: 02/28/2025]
Abstract
Glioblastoma represents the most aggressive type of brain cancer with minimal clinical advancements in recent decades attributed to the absence of efficient drug delivery strategies. In this study, we synthesized a series of vanadium complexes (V1-V4) and then constructed a lactoferrin (LF)-V4 nanoparticle (NP) delivery system. The nanoplatform crossed the blood-brain barrier by binding to low-density lipoprotein receptor-associated protein-1 and selectively targeted glioblastoma, ultimately inhibiting the growth of in situ glioblastoma tumors. LF-V4 NPs induced autophagic cell death in U87-MG cells by generating reactive oxygen species (ROS) that damaged the mitochondria. Further studies revealed that LF-V4 NPs triggered lipid peroxidation through the accumulation of ROS, the depletion of GSH, and the downregulation of GPX4 and SLC7A11, ultimately leading to ferroptosis in glioblastoma cells.
Collapse
Affiliation(s)
- Shuangshuang Gai
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
- Institute for History and Culture of Science and Technology, Guangxi Minzu University, Nanning 530006, China
| | - Qiwei Yan
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Shan Li
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Xuwei Zhong
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Yiming Qin
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| | - Ming Jiang
- School of Biological and Food Engineering, Guangxi Science and Technology Normal University, Laibin, Guangxi 546199, China
| |
Collapse
|
31
|
Liu J, Wang T, Dong J, Lu Y. The blood-brain barriers: novel nanocarriers for central nervous system diseases. J Nanobiotechnology 2025; 23:146. [PMID: 40011926 PMCID: PMC11866817 DOI: 10.1186/s12951-025-03247-8] [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: 11/16/2024] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
The central nervous system (CNS) diseases are major contributors to death and disability worldwide. However, the blood-brain barrier (BBB) often prevents drugs intended for CNS diseases from effectively crossing into the brain parenchyma to deliver their therapeutic effects. The blood-brain barrier is a semi-permeable barrier with high selectivity. The BBB primarily manages the transport of substances between the blood and the CNS. To enhance drug delivery for CNS disease treatment, various brain-based drug delivery strategies overcoming the BBB have been developed. Among them, nanoparticles (NPs) have been emphasized due to their multiple excellent properties. This review starts with an overview of the BBB's anatomical structure and physiological roles, and then explores the mechanisms, both endogenous and exogenous, that facilitate the NP passage across the BBB. The text also delves into how nanoparticles' shape, charge, size, and surface ligands affect their ability to cross the BBB and offers an overview of different nanoparticle classifications. This review concludes with an examination of the current challenges in utilizing nanomaterials for brain drug delivery and discusses corresponding directions for solutions. This review aims to propose innovative diagnostic and therapeutic approaches for CNS diseases and enhance drug design for more effective delivery across the BBB.
Collapse
Affiliation(s)
- Jiajun Liu
- State Key Laboratory of Green Biomanufacturing, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Tianjin Industrial Microbiology Key Laboratory, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ting Wang
- State Key Laboratory of Green Biomanufacturing, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| | - Jian Dong
- Tianjin Industrial Microbiology Key Laboratory, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yuan Lu
- State Key Laboratory of Green Biomanufacturing, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China.
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
32
|
Ahmed T, Alam KT. Biomimetic Nanoparticle Based Targeted mRNA Vaccine Delivery as a Novel Therapy for Glioblastoma Multiforme. AAPS PharmSciTech 2025; 26:68. [PMID: 39984771 DOI: 10.1208/s12249-025-03065-z] [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: 09/24/2024] [Accepted: 02/06/2025] [Indexed: 02/23/2025] Open
Abstract
The prognosis for patients with glioblastoma multiforme (GBM), an aggressive and deadly brain tumor, is poor due to the limited therapeutic options available. Biomimetic nanoparticles have emerged as a promising vehicle for targeted mRNA vaccine delivery, thanks to recent advances in nanotechnology. This presents a novel treatment method for GBM. This review explores the potential of using biomimetic nanoparticles to improve the specificity and effectiveness of mRNA vaccine against GBM. These nanoparticles can evade immune detection, cross the blood-brain barrier, & deliver mRNA directly to glioma cells by mimicking natural biological structures. This allows glioma cells to produce tumor-specific antigens that trigger strong immune responses against the tumor. This review discusses biomimetic nanoparticle design strategies, which are critical for optimizing transport and ensuring targeted action. These tactics include surface functionalization and encapsulation techniques. It also highlights the ongoing preclinical research and clinical trials that demonstrate the therapeutic advantages and challenges of this strategy. Biomimetic nanoparticles for mRNA vaccine delivery represent a new frontier in GBM treatment, which could impact the management of this deadly disease and improve patient outcomes by integrating cutting-edge nanotechnology with immunotherapy.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, School of Health and Life Sciences, North South University, Plot 15, Block B, Bashundhara R/A, Dhaka, 1229, Bangladesh.
| | - Kazi Tasnuva Alam
- Department of Pharmaceutical Sciences, School of Health and Life Sciences, North South University, Plot 15, Block B, Bashundhara R/A, Dhaka, 1229, Bangladesh
| |
Collapse
|
33
|
Grzegorzewski J, Michalak M, Wołoszczuk M, Bulicz M, Majchrzak-Celińska A. Nanotherapy of Glioblastoma-Where Hope Grows. Int J Mol Sci 2025; 26:1814. [PMID: 40076445 PMCID: PMC11898975 DOI: 10.3390/ijms26051814] [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: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Localization in the central nervous system, diffuse growth, the presence of stem cells, and numerous resistance mechanisms, all make glioblastoma (GBM) an incurable tumor. The standard treatment of GBM consisting of surgery; radio- and chemotherapy with temozolomide provides insufficient therapeutic benefit and needs to be updated with effective modern solutions. One of the most promising and intensively explored therapeutic approaches against GBM is the use of nanotherapy. The first, and so far only, nanoparticle-based therapy approved for GBM treatment is NanoThermTM. It is based on iron oxide nanoparticles and the thermal ablation of the tumor with a magnetic field. Numerous other types of nanotherapies are being evaluated, including polymer and lipid-based nanoformulations, nanodiscs, dendrimers, and metallic, silica, or bioderived nanoparticles, among others. The advantages of these nanoscale drug carriers include improved penetration across the blood-brain barrier, targeted drug delivery, biocompatibility, and lower systemic toxicity, while major problems with their implementation involve scaling up their production and high costs. Nevertheless, taking all the impressive benefits of nanotherapies into consideration, it seems obvious that the combined effort of the scientific world will need to be taken to tackle these challenges and implement these novel therapies into clinics, giving hope that the battle against GBM can finally be won.
Collapse
Affiliation(s)
- Jan Grzegorzewski
- The Student Scientific Society of Poznan University of Medical Sciences, 60-806 Poznań, Poland; (J.G.); (M.M.); (M.W.); (M.B.)
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| | - Maciej Michalak
- The Student Scientific Society of Poznan University of Medical Sciences, 60-806 Poznań, Poland; (J.G.); (M.M.); (M.W.); (M.B.)
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| | - Maria Wołoszczuk
- The Student Scientific Society of Poznan University of Medical Sciences, 60-806 Poznań, Poland; (J.G.); (M.M.); (M.W.); (M.B.)
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| | - Magdalena Bulicz
- The Student Scientific Society of Poznan University of Medical Sciences, 60-806 Poznań, Poland; (J.G.); (M.M.); (M.W.); (M.B.)
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| | - Aleksandra Majchrzak-Celińska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland
| |
Collapse
|
34
|
Agnihotri TG, Dahifale A, Gomte SS, Rout B, Peddinti V, Jain A. Nanosystems at Nexus: Navigating Nose-to-Brain Delivery for Glioblastoma Treatment. Mol Pharm 2025; 22:599-619. [PMID: 39746097 DOI: 10.1021/acs.molpharmaceut.4c00703] [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: 01/04/2025]
Abstract
Glioblastoma multiforme (GBM) is considered to be one of the most devastating brain tumors with a shorter life expectancy. Several factors contribute to the dismal prognosis of GBM patients including the complicated nature of GBM, the ability of tumor cells to resist treatment, and the difficulty of delivering drugs to the brain because of barriers like the blood-brain barrier (BBB) and blood-tumor barrier (BTB). The unique challenges posed by the BBB in delivering therapeutic agents to the brain have led to the development of innovative nanotechnology-based approaches. By exploiting the olfactory/trigeminal pathway, nanosystems offer a promising strategy for targeted drug delivery to the brain, glioblastoma tumors in particular. This review contemplates varied nanocarriers, including polymeric nanoparticles, lipid-based nanosystems, in situ gel formulations, peptide, and stem cell-based nanoformulations, signifying their utility in brain targeting with minimal systemic side effects. Emerging trends in gene therapy and immunotherapy in the context of GBM treatment have also been discussed. Since safety is a paramount aspect for any drug product to get approved, this review also delves into toxicological considerations associated with intranasal delivery of nanosystems. Regulatory aspects and critical factors for the successful development of intranasal products are also explored in this review. Overall, this review underscores the significant advancements in nanotechnology for nose-to-brain delivery and its potential impact on GBM management.
Collapse
Affiliation(s)
- Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar 382355, Gujarat, India
| | - Akanksha Dahifale
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar 382355, Gujarat, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar 382355, Gujarat, India
| | - Biswajit Rout
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar 382355, Gujarat, India
| | - Vasu Peddinti
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar 382355, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
35
|
Erickson NJ, Stavarache M, Tekedereli I, Kaplitt MG, Markert JM. Herpes Simplex Oncolytic Viral Therapy for Malignant Glioma and Mechanisms of Delivery. World Neurosurg 2025; 194:123595. [PMID: 39710201 PMCID: PMC12094189 DOI: 10.1016/j.wneu.2024.123595] [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: 09/25/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
The authors present a comprehensive review on the history and development of oncolytic herpes simplex viral therapies for malignant glioma with a focus on mechanisms of delivery in prior and ongoing clinical trials. This review highlights the advancements made with regard to delivering these therapies to a highly complex immunologic environment in the setting of the blood-brain and blood-tumor barrier in a safe and effective manner.
Collapse
Affiliation(s)
- Nicholas J Erickson
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mihaela Stavarache
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Ibrahim Tekedereli
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael G Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - James M Markert
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
36
|
Zhang G, Xu Y, Zhou A, Yu Y, Ning X, Bao H. Bioengineered NanoAid synergistically targets inflammatory pro-tumor processes to advance glioblastoma chemotherapy. NANOSCALE 2025; 17:2753-2768. [PMID: 39831463 DOI: 10.1039/d4nr04557b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Through transcriptomic analysis of patient-derived glioblastoma tissues, we identify an overactivation of inflammatory pathways that contribute to the development of a tumor-promoting microenvironment and therapeutic resistance. To address this critical mechanism, we present NanoAid, a biomimetic nanoplatform designed to target inflammatory pro-tumor processes to advance glioblastoma chemotherapy. NanoAid employs macrophage-membrane-liposome hybrids to optimize the delivery of COX-2 inhibitor parecoxib and paclitaxel. By inheriting macrophage characteristics, NanoAid not only efficiently traverses the blood-brain barrier and precisely accumulates within tumors but also enhances cancer cell uptake, thereby improving overall anticancer efficacy. Notably, the combination of parecoxib and paclitaxel effectively disrupts inflammatory pro-tumor processes while inducing a synergistic effect that inhibits tumor growth, overcomes therapeutic resistance, and minimizes adverse effects. This results in substantial tumor growth inhibition and extends the median survival of tumor-bearing mice. Thus, our study bridges clinical insights with fundamental research, potentially revolutionizing tumor therapy paradigms.
Collapse
Affiliation(s)
- Gui Zhang
- The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Yongle Yu
- Medical College of Guangxi University, Nanning 530004, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Hongguang Bao
- Department of Anaesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 211101, China.
| |
Collapse
|
37
|
Phon BWS, Chelliah SS, Osman DER, Bhuvanendran S, Radhakrishnan AK, Kamarudin MNA. Revisiting ABC Transporters and Their Clinical Significance in Glioblastoma. Pharmaceuticals (Basel) 2025; 18:102. [PMID: 39861164 PMCID: PMC11769420 DOI: 10.3390/ph18010102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Background: The multiple drug-resistant phenomenon has long since plagued the effectiveness of various chemotherapies used in the treatment of patients with glioblastoma (GBM), which is still incurable to this day. ATP-binding cassette (ABC) transporters function as drug transporters and have been touted to be the main culprits in developing resistance to xenobiotic drugs in GBM. Methods: This review systematically analyzed the efficacy of ABC transporters against various anticancer drugs from 16 studies identified from five databases (PubMed, Medline, Embase, Scopus, and ScienceDirect). Results: Inhibition of ABC transporters, especially ABCB1, improved drug efficacies. Staple GBM phenotypes, such as GBM stem cells and increased activation of the PI3K/Akt/NF-κB pathway, have been implicated in the expression of several ABC transporters. Using the datasets in The Cancer Genome Atlas and Gene Expression Omnibus, we found upregulated ABC transporters that either negatively impacted survival in univariate analyses (ABCA1, ABCA13, ABCB9, ABCD4) or were independent negative prognosis factors for patients with GBM (ABCA13, ABCB9). Our multivariate analysis further demonstrated three ABC transporters, ABCA13 (Hazard Ratio (HR) = 1.31, p = 0.017), ABCB9 (HR = 1.26, p = 0.03), and ABCB5 (HR = 0.77, p = 0.016), with the administration of alkylating agents (HR = 0.41, p < 0.001), were independent negative prognosis factors for patients with GBM. Conclusions: These findings reinforce the important role played by ABC transporters, particularly by ABCA13, ABCB9, and ABCB1, which could be potential targets that warrant further evaluations for alternate strategies to augment the effects of existing alkylating agents and xenobiotic drugs.
Collapse
Affiliation(s)
- Brandon Wee Siang Phon
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia; (B.W.S.P.); (S.S.C.); (D.E.-R.O.); (S.B.); (A.K.R.)
| | - Shalini Sundramurthi Chelliah
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia; (B.W.S.P.); (S.S.C.); (D.E.-R.O.); (S.B.); (A.K.R.)
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Dina El-Rabie Osman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia; (B.W.S.P.); (S.S.C.); (D.E.-R.O.); (S.B.); (A.K.R.)
| | - Saatheeyavaane Bhuvanendran
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia; (B.W.S.P.); (S.S.C.); (D.E.-R.O.); (S.B.); (A.K.R.)
| | - Ammu Kutty Radhakrishnan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia; (B.W.S.P.); (S.S.C.); (D.E.-R.O.); (S.B.); (A.K.R.)
| | - Muhamad Noor Alfarizal Kamarudin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia; (B.W.S.P.); (S.S.C.); (D.E.-R.O.); (S.B.); (A.K.R.)
| |
Collapse
|
38
|
Gahide G, Vendrell JF, Massicotte-Tisluck K, Caux S, Deschamps S, Noël-Lamy M, Belzile F, Roy LO, Fortin D. Safety of Cerebral Intra-Arterial Chemotherapy for the Treatment of Malignant Brain Tumours. J Clin Med 2025; 14:524. [PMID: 39860529 PMCID: PMC11766042 DOI: 10.3390/jcm14020524] [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: 11/18/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Cerebral intra-arterial chemotherapy (CIAC) has been demonstrated to achieve tumoricidal concentrations in cerebral tumour cells that are otherwise unachievable due to the presence of the blood-brain barrier. In this study, we sought to analyze the safety of CIAC in a cohort of patients treated at the Centre intégré universitaire de santé et de services sociaux de l'Estrie-Centre hospitalier universitaire de Sherbrooke (CIUSSS-CHUS). Methods: Treatments consisted of monthly CIAC. A neurological examination and neuroimaging study (MRI) were performed before every treatment. The files of patients enrolled in our CIAC programme were reviewed. Adverse events were analyzed and categorized. Results: Overall, 2991 CIAC procedures were performed in 642 patients. Pathologies were as follows: malignant gliomas (68.7%), cerebral metastasis (17.6%), and cerebral lymphomas (13.7%). Perfusion vessels were as follows: 80% internal carotid artery and 20% vertebral artery. The chemotherapeutic agents used were carboplatin (86.4%), methotrexate (28.5%), melphalan (28.6%), and liposomal doxorubicin (2.8%). Osmotic blood-brain barrier disruption (BBBD) was induced in 30.5% of treatments. Symptomatic vascular adverse events occurred during 27 procedures (0.9%) in 26 patients (4%). Namely, 23 strokes, one carotid artery occlusion (responsible for one of the strokes), and two intratumoral and one subdural hemorrhage. The absolute risk of stroke was 1.3% and 0.5% for CIAC with or without BBBD, respectively. The use of the vertebral artery significantly increased the risk of stroke. Drug infusion-related seizures occurred in 2.5% of patients; 83.8% were associated with methotrexate and 16.2% with carboplatin. Conclusions: CIAC is a safe procedure with a 0.9% overall rate of symptomatic complications (stroke, carotid occlusion, subdural hemorrhage or intratumoral bleeding-n = 27/2991) on a treatment basis, mainly consisting of strokes (85%, n = 23), with a modified NIH Stroke Scale score of 4.1 ± 3.3.
Collapse
Affiliation(s)
- Gérald Gahide
- Department of Medical Imaging, Division of Interventional Radiology, Centre Intégré Universitaire de Santé et de Services Sociaux de l’Estrie—Centre Hospitalier Universitaire de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada; (K.M.-T.); (S.C.); (S.D.); (M.N.-L.); (F.B.)
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Universitaire de Sherbrooke, 12e Avenue Nord, Porte 6, Sherbrooke, QC J1H 5N4, Canada;
| | - Jean-François Vendrell
- Institut de Cancérologie de Montpellier, Clinique de Val d’Aurelle, 34090 Montpellier, Cedex 5, France;
| | - Karine Massicotte-Tisluck
- Department of Medical Imaging, Division of Interventional Radiology, Centre Intégré Universitaire de Santé et de Services Sociaux de l’Estrie—Centre Hospitalier Universitaire de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada; (K.M.-T.); (S.C.); (S.D.); (M.N.-L.); (F.B.)
| | - Samuel Caux
- Department of Medical Imaging, Division of Interventional Radiology, Centre Intégré Universitaire de Santé et de Services Sociaux de l’Estrie—Centre Hospitalier Universitaire de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada; (K.M.-T.); (S.C.); (S.D.); (M.N.-L.); (F.B.)
- The Health Campus, Université de Sherbrooke, 3001 12e Avenue Nord, Immeuble X1, Sherbrooke, QC J1H 5N4, Canada
| | - Samuel Deschamps
- Department of Medical Imaging, Division of Interventional Radiology, Centre Intégré Universitaire de Santé et de Services Sociaux de l’Estrie—Centre Hospitalier Universitaire de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada; (K.M.-T.); (S.C.); (S.D.); (M.N.-L.); (F.B.)
- The Health Campus, Université de Sherbrooke, 3001 12e Avenue Nord, Immeuble X1, Sherbrooke, QC J1H 5N4, Canada
| | - Maxime Noël-Lamy
- Department of Medical Imaging, Division of Interventional Radiology, Centre Intégré Universitaire de Santé et de Services Sociaux de l’Estrie—Centre Hospitalier Universitaire de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada; (K.M.-T.); (S.C.); (S.D.); (M.N.-L.); (F.B.)
| | - François Belzile
- Department of Medical Imaging, Division of Interventional Radiology, Centre Intégré Universitaire de Santé et de Services Sociaux de l’Estrie—Centre Hospitalier Universitaire de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada; (K.M.-T.); (S.C.); (S.D.); (M.N.-L.); (F.B.)
| | - Laurent-Olivier Roy
- Department of Surgery, Division of Neurosurgery and Neuro-Oncology, Centre Intégré Universitaire de Santé et de Services Sociaux de l’Estrie—Centre Hospitalier Universitaire de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada;
| | - David Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Universitaire de Sherbrooke, 12e Avenue Nord, Porte 6, Sherbrooke, QC J1H 5N4, Canada;
- Department of Surgery, Division of Neurosurgery and Neuro-Oncology, Centre Intégré Universitaire de Santé et de Services Sociaux de l’Estrie—Centre Hospitalier Universitaire de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada;
| |
Collapse
|
39
|
Nolli MG, Terracciano M, Rea I, D'Errico S, Placido Mineo G, De Stefano L, Piccialli G, Riela S, Oliviero G, Borbone N. Mild-Temperature Catalyzed Hydrosilylation for Simplified Carbohydrate Functionalization of Porous Silicon Nanoparticles. Chemistry 2025; 31:e202402818. [PMID: 39679769 PMCID: PMC11724234 DOI: 10.1002/chem.202402818] [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: 07/26/2024] [Indexed: 12/17/2024]
Abstract
Porous silicon is one of the most explored nanostructured materials in various biomedical applications owing to its remarkable properties. However, its inherent chemical instability mandates a robust surface modification procedure, and proper surface bioengineering is essential to ensure its effectiveness in the biomedical field. In this study, we introduce a one-pot functionalization strategy that simultaneously stabilizes porous silicon nanoparticles and decorates their surface with carbohydrates through hydrosilylation chemistry, combining mild temperatures and a Lewis acid catalyst. This approach yielded a surface functionalization degree of 300 μmol g-1 in just 4 hours at 60 °C, significantly reducing both the prolonged reaction times and high temperatures typically associated with conventional hydrosilylation. Furthermore, this advancement opens the way for utilizing thermolabile molecules useful for surface bioengineering.
Collapse
Affiliation(s)
- Maria Grazia Nolli
- Department of PharmacyUniversity of Naples Federico IIvia D. Montesano 4980131NaplesItaly
| | - Monica Terracciano
- Department of PharmacyUniversity of Naples Federico IIvia D. Montesano 4980131NaplesItaly
| | - Ilaria Rea
- Naples Unit-National Research CouncilInstitute of Applied Sciences and Intelligent Systems (ISASI)via P. Castellino 11180131NaplesItaly
| | - Stefano D'Errico
- Department of PharmacyUniversity of Naples Federico IIvia D. Montesano 4980131NaplesItaly
| | | | - Luca De Stefano
- Naples Unit-National Research CouncilInstitute of Applied Sciences and Intelligent Systems (ISASI)via P. Castellino 11180131NaplesItaly
| | - Gennaro Piccialli
- Department of PharmacyUniversity of Naples Federico IIvia D. Montesano 4980131NaplesItaly
- ISBE-ITUniversity of Naples Federico IICorso Umberto I 4080138NaplesItaly
| | - Serena Riela
- Department of Chemical SciencesUniversity of CataniaVia A. Doria 695125CataniaItaly
| | - Giorgia Oliviero
- ISBE-ITUniversity of Naples Federico IICorso Umberto I 4080138NaplesItaly
- Department of Molecular Medicines and Medical BiotechnologiesUniversity of Naples Federico IIvia S. Pansini 580131NaplesItaly
| | - Nicola Borbone
- Department of PharmacyUniversity of Naples Federico IIvia D. Montesano 4980131NaplesItaly
- ISBE-ITUniversity of Naples Federico IICorso Umberto I 4080138NaplesItaly
| |
Collapse
|
40
|
Chiou J, Impedovo V, Huynh YB, Gorgoglione R, Penalva LOF, Lodi A, Brenner AJ, Tiziani S. Targeting Metabolic and Epigenetic Vulnerabilities in Glioblastoma with SN-38 and Rabusertib Combination Therapy. Int J Mol Sci 2025; 26:474. [PMID: 39859189 PMCID: PMC11764980 DOI: 10.3390/ijms26020474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Glioblastoma (GBM), the most prevalent primary malignant brain tumor, remains challenging to treat due to extensive inter- and intra-tumor heterogeneity. This variability demands combination treatments to improve therapeutic outcomes. A significant obstacle in treating GBM is the expression of O6-methylguanine-DNA methyltransferase, a DNA repair enzyme that reduces the efficacy of the standard alkylating agent, temozolomide, in about 50% of patients. This underscores the need for novel, more targeted therapies. Our study investigates the metabolic-epigenetic impact of combining SN-38, a novel topoisomerase inhibitor inducing DNA double-strand breaks, with rabusertib, a checkpoint kinase 1 inhibitor. We identified this synergistic combination through high-throughput drug screening across a panel of GBM cell lines using a cancer drug library combined with SN-38. A secondary metabolic screening with the PEDS algorithm demonstrated a synergistic modulation of purine, one-carbon, and redox metabolism. Furthermore, the combined treatment led to the significant depletion of epigenetically relevant metabolites such as 5-methyl-cytosine, acetyl-lysine, and trimethyl-lysine. Reduced intermediates of the glutathione cycle indicated increased cellular stress following combinatorial treatment. Overall, the combination of SN-38 and rabusertib synergistically disrupts metabolites associated with epigenetic adaptations, leading to cytotoxicity independent of O6-methylguanine-DNA methyltransferase status, thereby underpinning this combination as a promising candidate for combinatorial therapy in GBM.
Collapse
Affiliation(s)
- Jennifer Chiou
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (J.C.); (V.I.); (Y.B.H.); (R.G.); (A.L.)
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| | - Valeria Impedovo
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (J.C.); (V.I.); (Y.B.H.); (R.G.); (A.L.)
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| | - Yen Bao Huynh
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (J.C.); (V.I.); (Y.B.H.); (R.G.); (A.L.)
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| | - Ruggiero Gorgoglione
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (J.C.); (V.I.); (Y.B.H.); (R.G.); (A.L.)
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| | - Luiz O. F. Penalva
- Children’s Cancer Research Institute, UT Health San Antonio, San Antonio, TX 78229, USA;
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Alessia Lodi
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (J.C.); (V.I.); (Y.B.H.); (R.G.); (A.L.)
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| | - Andrew J. Brenner
- Mays Cancer Center, UT Health San Antonio, 7979 Wurzbach Road, San Antonio, TX 78229, USA;
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA; (J.C.); (V.I.); (Y.B.H.); (R.G.); (A.L.)
- Dell Pediatric Research Institute, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, USA
| |
Collapse
|
41
|
Mokarram N, Case A, Hossainy NN, Lyon JG, MacDonald TJ, Bellamkonda R. Device-assisted strategies for drug delivery across the blood-brain barrier to treat glioblastoma. COMMUNICATIONS MATERIALS 2025; 6:5. [PMID: 39790893 PMCID: PMC11706785 DOI: 10.1038/s43246-024-00721-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025]
Abstract
The blood-brain barrier, essential for protecting the central nervous system, also restricts drug delivery to this region. Thus, delivering drugs across the blood-brain barrier is an active research area in immunology, oncology, and neurology; moreover, novel methods are urgently needed to expand therapeutic options for central nervous system pathologies. While previous strategies have focused on small molecules that modulate blood-brain barrier permeability or penetrate the barrier, there is an increased focus on biomedical devices-external or implanted-for improving drug delivery. Here, we review device-assisted drug delivery across the blood-brain barrier, emphasizing its application in glioblastoma, an aggressively malignant primary brain cancer in which the blood-brain barrier plays a central role. We examine the blood-brain barrier and its features in glioblastoma, emerging models for studying the blood-brain barrier, and device-assisted methods for crossing the blood-brain barrier. We conclude by presenting methods to monitor the blood-brain barrier and paradigms for combined cross-BBB drug delivery.
Collapse
Affiliation(s)
- Nassir Mokarram
- Department of Neurosurgery, Emory University, Atlanta, GA USA
| | - Ayden Case
- Trinity College of Arts and Sciences, Duke University, Durham, NC USA
| | | | - Johnathan G. Lyon
- Department of Biomedical Engineering, Duke University, Durham, NC USA
| | - Tobey J. MacDonald
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA USA
| | | |
Collapse
|
42
|
Mousavi MA, Rezaei M, Pourhamzeh M, Salari M, Hossein-Khannazer N, Shpichka A, Nabavi SM, Timashev P, Vosough M. Translational Approach using Advanced Therapy Medicinal Products for Huntington's Disease. Curr Rev Clin Exp Pharmacol 2025; 20:14-31. [PMID: 38797903 DOI: 10.2174/0127724328300166240510071548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/02/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024]
Abstract
Current therapeutic approaches for Huntington's disease (HD) focus on symptomatic treatment. Therefore, the unavailability of efficient disease-modifying medicines is a significant challenge. Regarding the molecular etiology, targeting the mutant gene or advanced translational steps could be considered promising strategies. The evidence in gene therapy suggests various molecular techniques, including knocking down mHTT expression using antisense oligonucleotides and small interfering RNAs and gene editing with zinc finger proteins and CRISPR-Cas9-based techniques. Several post-transcriptional and post-translational modifications have also been proposed. However, the efficacy and long-term side effects of these modalities have yet to be verified. Currently, cell therapy can be employed in combination with conventional treatment and could be used for HD in which the structural and functional restoration of degenerated neurons can occur. Several animal models have been established recently to develop cell-based therapies using renewable cell sources such as embryonic stem cells, induced pluripotent stem cells, mesenchymal stromal cells, and neural stem cells. These models face numerous challenges in translation into clinics. Nevertheless, investigations in Advanced Therapy Medicinal Products (ATMPs) open a promising window for HD research and their clinical application. In this study, the ATMPs entry pathway in HD management was highlighted, and their advantages and disadvantages were discussed.
Collapse
Affiliation(s)
- Maryam Alsadat Mousavi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maliheh Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Departments of Pathology and Medicine, UC San Diego, La Jolla, CA, USA
| | - Mehri Salari
- Department of Neurology, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare," Sechenov University, Moscow, Russia
| | - Seyed Massood Nabavi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- World-Class Research Center "Digital Biodesign and Personalized Healthcare," Sechenov University, Moscow, Russia
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 141-83 Stockholm, Sweden
| |
Collapse
|
43
|
Wahengbam GS, Nirmal S, Nandwana J, Kar S, Kumari V, Mishra R, Singh A. Polymeric Nanoparticles Revolutionizing Brain Cancer Therapy: A Comprehensive Review of Strategies and Advances. Crit Rev Ther Drug Carrier Syst 2025; 42:73-106. [PMID: 39819464 DOI: 10.1615/critrevtherdrugcarriersyst.2024051822] [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: 01/19/2025]
Abstract
Brain cancer continues to be one of the most formidable malignancies to manage, mainly attributable to the presence of the blood-brain barrier (BBB) limiting the permeability of drugs and the diverse characteristics of brain tumors complicating treatment. The management of brain tumors has been hampered by many different factors, including the impermeability of the BBB, which restricts the delivery of chemotherapeutic agents to the tumor site, as well as intertumoral heterogeneity and the influence of brain tumor stem cells. In addition, small molecular weight drugs cannot specifically accumulate in malignant cells and have a limited circulation half-life. Nanoparticles (NPs) can be engineered to traverse the BBB and transport therapeutic medications directly into the brain, enhancing their efficacy compared with the conventional delivery of unbound drugs. Surface modifications of NPs can boost their efficiency by increasing their selectivity towards tumor receptors. This review covers treatment methods for malignant gliomas, associated risk factors, and improvements in brain drug administration, emphasizing the future potential of polymeric NPs and their mechanism for crossing the BBB. To surmount these obstacles, the newly formulated drug-delivery approach utilizing NPs, particularly those coated with cell membranes, has demonstrated potential in treating brain cancer. These NPs provide targeted tumor specificity, biocompatibility, extended circulation, enhanced BBB penetration, and immune evasion. This review focuses on coating strategies for PLGA NPs, particularly dual-targeting methods, to enhance BBB permeability and tumor-targeted delivery of drugs in brain cancer.
Collapse
Affiliation(s)
| | - Sakshi Nirmal
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Jai Nandwana
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Swatileena Kar
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Vandana Kumari
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Rajeev Mishra
- Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, India
| | | |
Collapse
|
44
|
Dhiman A, Rana D, Benival D, Garkhal K. Comprehensive insights into glioblastoma multiforme: drug delivery challenges and multimodal treatment strategies. Ther Deliv 2025; 16:87-115. [PMID: 39445563 PMCID: PMC11703381 DOI: 10.1080/20415990.2024.2415281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and malignant brain tumors, with a high prevalence in elderly population. Most chemotherapeutic agents fail to reach the tumor site due to various challenges. However, smart nanocarriers have demonstrated excellent drug-loading capabilities, enabling them to cross the blood brain tumor barrier for the GBM treatment. Surface modification of nanocarriers has significantly enhanced their potential for targeting therapeutics. Moreover, recent innovations in drug therapies, such as the incorporation of theranostic agents in nanocarriers and antibody-drug conjugates, have offered newer insights for both diagnosis and treatment. This review focuses on recent advances in new therapeutic interventions for GBM, with an emphasis on the nanotheranostics systems to maximize therapeutic and diagnostic outcomes.
Collapse
Affiliation(s)
- Ashish Dhiman
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Dhwani Rana
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Derajram Benival
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| | - Kalpna Garkhal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research-Ahmedabad (NIPER-A), Gandhinagar, 382355, Gujarat, India
| |
Collapse
|
45
|
Ambele MA, Maebele LT, Mulaudzi TV, Kungoane T, Damane BP. Advances in nano-delivery of phytochemicals for glioblastoma treatment. DISCOVER NANO 2024; 19:216. [PMID: 39718730 DOI: 10.1186/s11671-024-04172-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Glioblastoma (GBM) is an aggressive brain tumor characterized by cellular and molecular diversity. This diversity presents significant challenges for treatment and leads to poor prognosis. Surgery remains the primary treatment of choice for GBMs, but it often results in tumor recurrence due to complex interactions between GBM cells and the peritumoral brain zone. Phytochemicals have shown promising anticancer activity in in-vitro studies and are being investigated as potential treatments for various cancers, including GBM. However, some phytochemicals have failed to translate their efficacy to pre-clinical studies due to limited penetration into the tumor microenvironment, leading to high toxicity. Thus, combining phytochemicals with nanotechnology has emerged as a promising alternative for treating GBM. This review explores the potential of utilizing specific nanoparticles to deliver known anticancer phytochemicals directly to tumor cells. This method has demonstrated potential in overcoming the challenges of the complex GBM microenvironment, including the tight blood-brain barrier while minimizing damage to healthy brain tissue. Therefore, employing this interdisciplinary approach holds significant promise for developing effective phyto-nanomedicines for GBM and improving patient outcomes.
Collapse
Affiliation(s)
- Melvin Anyasi Ambele
- Department of Oral and Maxillofacial Pathology, Faculty of Health Sciences, School of Dentistry, University of Pretoria, P.O. Box 1266, Pretoria, 0001, South Africa.
- Department of Immunology, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, P.O. Box 0084, Gezina, South Africa.
| | - Lorraine Tshegofatso Maebele
- Department of Surgery, Level 7, Bridge E, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa
| | - Thanyani Victor Mulaudzi
- Department of Surgery, Level 7, Bridge E, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa
| | - Tsholofelo Kungoane
- Department of Oral and Maxillofacial Pathology, Faculty of Health Sciences, School of Dentistry, University of Pretoria, P.O. Box 1266, Pretoria, 0001, South Africa
| | - Botle Precious Damane
- Department of Surgery, Level 7, Bridge E, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Private Bag X323, Arcadia, 0007, South Africa.
| |
Collapse
|
46
|
Lei K, Sheng Y, Li Y, Zhou Z, Zhu X, Huang K. Dynamic Detection of the E3-PROTAC-Target Protein Ternary Complex In Vitro and In Vivo via Bimolecular Fluorescence Complementation. ACS OMEGA 2024; 9:49739-49748. [PMID: 39713624 PMCID: PMC11656243 DOI: 10.1021/acsomega.4c08186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) have played an important role in the development of protein-targeted degradation drugs. However, effective tools are urgently required for the further development and validation of PROTACs. We developed a high-potency reporter (AKT-PROTAC-Reporter; APR) for PROTACs that specifically targets AKT. The APR successfully detected the status and levels of the AKT-PROTAC-CRBN ternary complex in vivo and in vitro. The APR is based on a bimolecular fluorescence complementation system, where EGFP and luciferase were used as reporter signals for in vitro and in vivo experiments, respectively, with remarkable success. The absence of E3 ligase ubiquitin recruitment activity in the APR can significantly improve the reporting performance of the APR; however, this results in difficulties in the detection of the degradation efficiency of PROTAC target proteins. Our results show that the APR can sensitively, quickly, and effectively detect the presence of terpolymers. Furthermore, the APR can determine the specificity and degradation efficiency of the PROTAC via a fluorescence signal or bioluminescence signal intensity and can efficiently screen PROTACs for a certain target protein.
Collapse
Affiliation(s)
- Kunjian Lei
- Department
of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Institute
of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Yilei Sheng
- Department
of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Institute
of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Yishuang Li
- Department
of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Institute
of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Zhihong Zhou
- Department
of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Institute
of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Xingen Zhu
- Department
of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Institute
of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| | - Kai Huang
- Department
of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
- Institute
of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, P. R. China
| |
Collapse
|
47
|
Li Y, Wu C, Long X, Wang X, Gao W, Deng K, Xie B, Zhang S, Wu M, Liu Q. Single-cell transcriptomic analysis of glioblastoma reveals pericytes contributing to the blood-brain-tumor barrier and tumor progression. MedComm (Beijing) 2024; 5:e70014. [PMID: 39640361 PMCID: PMC11617595 DOI: 10.1002/mco2.70014] [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: 12/16/2023] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 12/07/2024] Open
Abstract
The blood-brain barrier is often altered in glioblastoma (GBM) creating a blood-brain-tumor barrier (BBTB) composed of pericytes. The BBTB affects chemotherapy efficacy. However, the expression signatures of BBTB-associated pericytes remain unclear. We aimed to identify BBTB-associated pericytes in single-cell RNA sequencing data of GBM using pericyte markers, a normal brain pericyte expression signature, and functional enrichment. We identified parathyroid hormone receptor-1 (PTH1R) as a potential marker of pericytes associated with BBTB function. These pericytes interact with other cells in GBM mainly through extracellular matrix-integrin signaling pathways. Compared with normal pericytes, pericytes in GBM exhibited upregulation of several ECM genes (including collagen IV and FN1), and high expression levels of these genes were associated with a poor prognosis. Cell line experiments showed that PTH1R knockdown in pericytes increased collagen IV and FN1 expression levels. In mice models, the expression levels of PTH1R, collagen IV, and FN1 were consistent with these trends. Evans Blue leakage and IgG detection in the brain tissue suggested a negative correlation between PTH1R expression levels and blood-brain barrier function. Further, a risk model based on differentially expressed genes in PTH1R+ pericytes had predictive value for GBM, as validated using independent and in-house cohorts.
Collapse
Affiliation(s)
- Yuzhe Li
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of NeurosurgeryChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Changwu Wu
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xinmiao Long
- Cancer Research InstituteCentral South UniversityChangshaHunanChina
| | - Xiangyu Wang
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Wei Gao
- Cancer Research InstituteCentral South UniversityChangshaHunanChina
| | - Kun Deng
- Cancer Research InstituteCentral South UniversityChangshaHunanChina
| | - Bo Xie
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Sen Zhang
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Minghua Wu
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- Cancer Research InstituteCentral South UniversityChangshaHunanChina
| | - Qing Liu
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
48
|
Docrat TF, Eltahir AOE, Hussein AA, Marnewick JL. Green synthesis of metal nanocarriers: A perspective for targeting glioblastoma. Drug Discov Today 2024; 29:104219. [PMID: 39476945 DOI: 10.1016/j.drudis.2024.104219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/05/2024] [Accepted: 10/24/2024] [Indexed: 11/11/2024]
Abstract
Glioblastoma, the most aggressive brain cancer, is challenging to treat owing to the difficulty of crossing the blood-brain barrier, high recurrence rates and significant mortality. This review highlights the potential of green synthesis methods in developing metal nanoparticles (MNPs) as a sustainable solution for drug delivery systems targeting glioblastoma. We explore the unique properties and modes of action of MNPs synthesised through eco-friendly processes by focusing on their bioavailability and precision in brain targeting, and discuss the potential of MNPs to target glioblastoma at the molecular level. Integrating green synthesis into cancer therapeutics represents a novel paradigm shift towards treatments with higher efficacy and lower environmental impact, offering hope in the fight against glioblastoma.
Collapse
Affiliation(s)
- Taskeen F Docrat
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville 7535, South Africa.
| | - Ali O E Eltahir
- Department of Chemistry, Cape Peninsula University of Technology, Bellville 7535, South Africa; Permanent address: Department of Chemistry, Omdurman Islamic University, Omdurman, P.O. Box 382, Khartoum, Sudan
| | - Ahmed A Hussein
- Department of Chemistry, Cape Peninsula University of Technology, Bellville 7535, South Africa
| | - Jeanine L Marnewick
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville 7535, South Africa
| |
Collapse
|
49
|
Wang M, Bergès R, Malfanti A, Préat V, Bastiancich C. Local delivery of doxorubicin prodrug via lipid nanocapsule-based hydrogel for the treatment of glioblastoma. Drug Deliv Transl Res 2024; 14:3322-3338. [PMID: 37889402 PMCID: PMC11499358 DOI: 10.1007/s13346-023-01456-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2023] [Indexed: 10/28/2023]
Abstract
Glioblastoma (GBM) recurrences appear in most cases around the resection cavity borders and arise from residual GBM cells that cannot be removed by surgery. Here, we propose a novel treatment that combines the advantages of nanomedicine and local drug delivery to target these infiltrating GBM cells. We developed an injectable lipid nanocapsule (LNC)-based formulation loaded with lauroyl-doxorubicin prodrug (DOXC12). Firstly, we demonstrated the efficacy of intratumoral administration of DOXC12 in GL261 GBM-bearing mice, which extended mouse survival. Then, we formulated an injectable hydrogel by mixing the appropriate amount of prodrug with the lipophilic components of LNC. We optimized the hydrogel by incorporating cytidine-C16 (CytC16) to achieve a mechanical stiffness adapted for an application in the brain post-surgery (DOXC12-LNCCL). DOXC12-LNCCL exhibited high DOXC12 encapsulation efficiency (95%) and a size of approximately 60 nm with sustained drug release for over 1 month in vitro. DOXC12-LNCCL exhibited enhanced cytotoxicity compared to free DOXC12 (IC50 of 349 and 86 nM, respectively) on GL261 GBM cells and prevented the growth of GL261 spheroids cultured on organotypic brain slices. In vivo, post-surgical treatment with DOXC12-LNCCL significantly improved the survival of GL261-bearing mice. The combination of this local treatment with the systemic administration of anti-inflammatory drug ibuprofen further delayed the onset of recurrences. In conclusion, our study presents a promising therapeutic approach for the treatment of GBM. By targeting residual GBM cells and reducing the inflammation post-surgery, we present a new strategy to delay the onset of recurrences in the gap period between surgery and standard of care therapy.
Collapse
Affiliation(s)
- Mingchao Wang
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium
| | - Raphaël Bergès
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 27 Boulevard Jean Moulin, Marseille, 13005, France
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium.
| | - Chiara Bastiancich
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium.
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 27 Boulevard Jean Moulin, Marseille, 13005, France.
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, Turin, 10125, Italy.
| |
Collapse
|
50
|
Jourdain MA, Eyer J. Recent advances in liposomes and peptide-based therapeutics for glioblastoma treatment. J Control Release 2024; 376:732-752. [PMID: 39437968 DOI: 10.1016/j.jconrel.2024.10.037] [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: 02/07/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
In the context of glioblastoma treatment, the penetration of drugs is drastically limited by the blood-brain-barrier (BBB). Emerging therapies have focused on the field of therapeutic peptides for their excellent BBB targeting properties that promote a deep tumor penetration. Peptide-based strategies are also renowned for their abilities of driving cargo such as liposomal system allowing an active targeting of receptors overexpressed on GBM cells. This review provides a detailed description of the internalization mechanisms of specific GBM homing and penetrating peptides as well as the latest in vitro/in vivo studies of liposomes functionalized with them. The purpose of this review is to summarize a selection of promising pre-clinical results that demonstrate the advantages of this nanosystem, including an increase of tumor cell targeting, triggering drug accumulation and thus a strong antitumor effect. Aware of the early stage of these studies, many challenges need to be overcome to promote peptide-directed liposome at clinical level. In particular, the lack of suitable production, the difficulty to characterize the nanosystem and therapeutic competition leaded by antibodies.
Collapse
Affiliation(s)
- M-A Jourdain
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France.
| | - J Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| |
Collapse
|