1
|
Méndez-Luna D, Morelos-Garnica LA, García-Sánchez JR, Joucla G, Bonneau L, Bakalara N, Correa-Basurto J. Preclinical approach of two novel tetrahydroquinoline derivatives targeting GPER and Bcl-2 for anti-glioblastoma therapy. Sci Rep 2025; 15:17710. [PMID: 40399430 PMCID: PMC12095820 DOI: 10.1038/s41598-025-02186-6] [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: 12/20/2024] [Accepted: 05/12/2025] [Indexed: 05/23/2025] Open
Abstract
Glioblastoma multiforme (GBM), is a rapidly growing and aggressive brain tumor that can arise de novo in the brain or evolve from lower-grade astrocytoma. This malignancy represents a medical challenge due to the tumor´s localization in the brain, high rates of Temozolomide (TMZ) resistance, and extensive malignant cell parenchymal infiltration, among other factors. Consequently, new drug discovery efforts have focused on targeting pivotal pharmacological targets such as GPER and Bcl-2, presenting a promising strategy for developing new GBM treatments. Herein, we present the results of an improved structure guided design of anti-glioblastoma compounds, L-06 and L-37, both containing the tetrahydroquinoline scaffold and a sulfonamide moiety recognized by GPER and Bcl-2 binding sites, respectively. Both compounds were evaluated in a battery of in vitro assays to measure their anti-glioblastoma activity. L-06 and L-37 were subjected to chemical stability testing under forced degradation conditions indicated minimal degradation from 0.15 to 13.6%. Additionally, antiproliferative evaluation in 2D cell culture yielded IC50 values between 39 and 67 µM in GBM cell lines LN18 and U373, consistent with Gossypol, a well-known Bcl-2 inhibitor. G-15 and L-37 to a greater extent than L-06, inhibit neurospheres formation in glioblastoma stem cells (Gli4) cultured in a proliferation medium. Moreover, in 3D antiproliferative assays using glioblastoma stem cells on non-aligned nanofibers L-37 showed better inhibitory effect than L-06. Interestingly, L-06 than L-37 exhibited an antimigratory effect on glioblastoma stem cells loaded onto aligned nanofibers at concentrations where no antiproliferative activity were observed, unlike G-15, a poorly water soluble GPER antagonist. Collectively, these findings establish a preclinical foundation for L-37 and L-06 as potential anti-glioblastoma agents and support their further investigation as therapeutic candidates.
Collapse
Affiliation(s)
- David Méndez-Luna
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Alcaldía Miguel Hidalgo, Mexico City, C.P. 11340, México
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Zacatenco, Av. Wilfrido Massieu 399, Col. Nueva Industrial Vallejo, Alcaldía Gustavo A. Madero, Ciudad de México, C.P. 07738, México
| | - Loreley-Araceli Morelos-Garnica
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Alcaldía Miguel Hidalgo, Mexico City, C.P. 11340, México
| | - José-Rubén García-Sánchez
- Laboratorio de Oncología Molecular y Estrés Oxidativo de la Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, s/n, Col. Casco de Santo Tomas, Ciudad de México, 11340, Mexico
| | - Gilles Joucla
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, F-33600, France
| | - Laurent Bonneau
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, F-33600, France
| | - Norbert Bakalara
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, F-33600, France.
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Alcaldía Miguel Hidalgo, Mexico City, C.P. 11340, México.
| |
Collapse
|
2
|
Wang Q, Zhang Y, Ma K, Lin P, Wang Y, Wang R, Li H, Li Z, Wang G. Plexin B2 in physiology and pathophysiology of the central nervous system. Int Immunopharmacol 2025; 155:114627. [PMID: 40220620 DOI: 10.1016/j.intimp.2025.114627] [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: 02/10/2025] [Revised: 04/05/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
The Plexin protein family was initially found in 1995, comprising subfamilies from Plexin A to Plexin D. Plexin B2, a member of the Plexin subfamily, has widespread expression in many human organs and tissues, particularly in the nervous system where expression levels are significantly heightened. The biological roles of Plexin B2 are mostly determined by its protein structure and functional domains. These domains regulate the binding selectivity and affinity for ligands. Ligand binding activates signal transduction pathways, resulting in regulatory effects on several biological processes. This includes managing brain growth and change, keeping angiogenesis and vascular homeostasis in check, and preventing the start, growth, and metastasis of cancer. Plexin B2 has also been associated with the onset of many nervous system illnesses. Plexin B2 aids in the invasion and spread of malignant cells, facilitates nerve healing following spinal cord damage, and plays a role in the etiology of schizophrenia. This article thoroughly examines the existing research on Plexin B2 and its importance in central nervous system biology. Simultaneously, it investigates the regulatory function of Plexin B2 across many cell types in the central nervous system, specifically neural stem cells, neurons, microglia, and astrocytes. This study examines the current knowledge of Plexin B2's role in central nervous system diseases, including schizophrenia, spinal cord injury, neuroblastoma, and fear memory. Overall, the prospects for the clinical translation of Plexin B2 are promising. However, challenges related to specificity and drug delivery must be addressed. Future research could explore the integration of nanodrug delivery systems to enhance the clinical application of Plexin B2-targeted therapies.
Collapse
Affiliation(s)
- Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yuan Zhang
- Department of Pharmacy, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai 200120, China
| | - Kaixuan Ma
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Peng Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Ran Wang
- School of Pharmacy, Harbin Medical University, Daqing, Heilongjiang 163319, China
| | - He Li
- Department of Parasitology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China.
| | - Guangtian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China; Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
3
|
Bhutani B, Sharma V, Ganguly NK, Rana R. Unravelling the modified T cell receptor through Gen-Next CAR T cell therapy in Glioblastoma: Current status and future challenges. Biomed Pharmacother 2025; 186:117987. [PMID: 40117901 DOI: 10.1016/j.biopha.2025.117987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025] Open
Abstract
PURPOSE Despite current technological advancements in the treatment of glioma, immediate alleviation of symptoms can be catered by therapeutic modalities, including surgery, chemotherapy, and combinatorial radiotherapy that exploit aberrations of glioma. Additionally, a small number of target antigens, their heterogeneity, and immune evasion are the potential reasons for developing targeted therapies. This oncologic milestone has catalyzed interest in developing immunotherapies against Glioblastoma to improve overall survival and cure patients with high-grade glioma. The next-gen CAR-T Cell therapy is one of the effective immunotherapeutic strategies in which autologous T cells have been modified to express receptors against GBM and it modulates cytotoxicity. METHODS In this review article, we examine preclinical and clinical outcomes, and limitations as well as present cutting-edge techniques to improve the function of CAR-T cell therapy and explore the possibility of combination therapy. FINDINGS To date, several CAR T-cell therapies are being evaluated in clinical trials for GBM and other brain malignancies and multiple preclinical studies have demonstrated encouraging outcomes. IMPLICATIONS CAR-T cell therapy represents a promising therapeutic paradigm in the treatment of solid tumors but a few limitations include, the blood-brain barrier (BBB), antigen escape, tumor microenvironment (TME), tumor heterogeneity, and its plasticity that suppresses immune responses weakens the ability of this therapy. Additional investigation is required that can accurately identify the targets and reflect the similar architecture of glioblastoma, thus optimizing the efficiency of CAR-T cell therapy; allowing for the selection of patients most likely to benefit from immuno-based treatments.
Collapse
Affiliation(s)
- Bhavya Bhutani
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Vyoma Sharma
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India.
| |
Collapse
|
4
|
Abbasian P, Ryner L, McCurdy B, Kakumanu S, Essig M, Venugopal N, Guan J, Pitz M. Predicting brain tumour growth patterns using a novel MRI-based tumour spread map: application to radiotherapy planning. Med Phys 2025; 52:2909-2921. [PMID: 39865295 DOI: 10.1002/mp.17640] [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: 01/05/2024] [Revised: 08/05/2024] [Accepted: 12/22/2024] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND The treatment of glioblastomas (GBM) with radiation therapy is extremely challenging due to their invasive nature and high recurrence rate within normal brain tissue. PURPOSE In this work, we present a new metric called the tumour spread (TS) map, which utilizes diffusion tensor imaging (DTI) to predict the probable direction of tumour cells spread along fiber tracts. We hypothesized that the TS map could serve as a predictive tool for identifying patterns of likely recurrence in patients with GBM and, therefore, be used to modify the delivery of radiation treatment to pre-emptively target regions at high risk of tumour spread. METHODS In this proof-of-concept study, we visualized the white matter fiber tract pathways within the brain using diffusion tensor tractography and developed an algorithm which mathematically calculates a relative probability index in each voxel, resulting in the generation of the TS map. Based on the information provided by the TS map, the original radiotherapy target volume was then modified to include areas with a higher probability of tumour spread and exclude other areas with a lower probability of spread. A volumetric modulated arc therapy (VMAT) treatment plan was then developed utilizing the modified target volumes and subsequently compared to that using the original target volumes. Follow-up anatomical imaging obtained 8 months post-surgery was assessed to validate our findings. RESULTS A TS map was generated on a glioblastoma patient demonstrating a relative probability of tumour spread along fiber tracts throughout the brain. The modified planning target volume better covered brain regions with a higher risk of tumour spread while still demonstrating a 21% reduction in volume compared to the original planning target volume, resulting in greater preservation of normal tissue. The modified VMAT plan resulted in an average mean dose to four identified recurrences of 80% of the prescription dose, while the original VMAT plan delivered only 63% of the prescription dose as the average mean dose to the recurrences. CONCLUSION The utilization of tractography and the generation of corresponding TS maps offer a promising approach to predicting patterns of tumour recurrence and optimizing treatment delivery. Further research is needed to validate the predictive value of the TS map on a larger cohort of patients and explore its potential in personalized treatment strategies for GBM patients.
Collapse
Affiliation(s)
- Parandoush Abbasian
- Physics and Astronomy, University of Manitoba, Winnipeg, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, Canada
- Medical Physics, CancerCare Manitoba, Winnipeg, Canada
| | - Lawrence Ryner
- Physics and Astronomy, University of Manitoba, Winnipeg, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, Canada
- Medical Physics, CancerCare Manitoba, Winnipeg, Canada
| | - Boyd McCurdy
- Physics and Astronomy, University of Manitoba, Winnipeg, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, Canada
- Medical Physics, CancerCare Manitoba, Winnipeg, Canada
| | - Saranya Kakumanu
- Radiology, University of Manitoba, Winnipeg, Canada
- Radiation Oncology, CancerCare Manitoba, Winnipeg, Canada
| | - Marco Essig
- Department of Radiology, Winnipeg Health Sciences Centre, Winnipeg, Canada
| | - Niranjan Venugopal
- Physics and Astronomy, University of Manitoba, Winnipeg, Canada
- Medical Physics, CancerCare Manitoba, Winnipeg, Canada
| | - James Guan
- Medical Physics, CancerCare Manitoba, Winnipeg, Canada
| | - Marshall Pitz
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, Canada
- Internal Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
5
|
Zhong R, He H, Wang X. Novel neutrophil targeting platforms in treating Glioblastoma: Latest evidence and therapeutic approaches. Int Immunopharmacol 2025; 150:114173. [PMID: 39938169 DOI: 10.1016/j.intimp.2025.114173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/14/2025]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal type of primary brain tumor, characterized by its rapid growth, resistance to conventional therapies, and a highly immunosuppressive tumor microenvironment (TME). Recent studies have highlighted the critical role of neutrophils in the progression of GBM, where they contribute to tumor growth, invasion, and treatment resistance. As a result, neutrophils have emerged as a promising target for therapeutic intervention in GBM. Various strategies are being investigated to specifically target neutrophils within the GBM environment, including using small molecules, antibodies, and nanoparticle-based methods. These approaches aim to regulate neutrophils' recruitment, activation, and functions. This study reviews the latest findings regarding the involvement of neutrophils in GBM, explores potential techniques targeting neutrophils for therapeutic purposes, and discusses current clinical studies and prospects in this rapidly evolving field. By studying the diverse functions of neutrophils in GBM, these innovative therapeutic strategies can help address some of the most significant challenges in treating this malignancy.
Collapse
Affiliation(s)
- Rui Zhong
- Department of Neurosurgery, The First People's Hospital of Lin'an District, Hangzhou 311300, China
| | - Hongmei He
- Department of Neurosurgery, The First People's Hospital of Lin'an District, Hangzhou 311300, China
| | - Xiande Wang
- Department of Neurosurgery, The First People's Hospital of Lin'an District, Hangzhou 311300, China.
| |
Collapse
|
6
|
Ghosh SR, Lally AR, Pecorari IL, Reynolds J, Ledet A, Begley S, Diaz EJ, Zhu E, Joseph K, MPhil KM, Schulder M, Johanns T, Ziemba YC, Agarwal V. The impact of sociodemographic background on clinical presentation of high-grade gliomas: a multi-institutional retrospective analysis. J Neurooncol 2025:10.1007/s11060-025-05012-1. [PMID: 40131577 DOI: 10.1007/s11060-025-05012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE High-grade gliomas (HGG; WHO III/IV) are among the most devastating intracranial malignancies, and outcomes may be associated with demographic, biological and environmental factors. Although research exists on the association of sociodemographic background with outcomes, the literature lacks data on the effect of sociodemographic background on clinical presentation. In this study, we aimed to examine race- and ethnicity-related differences in HGG presentation and diagnosis. METHODS We conducted a chart review of patients treated for HGG between 2015 and 2021 at three high-volume academic medical centers. A total of 314 patients were analyzed. 173 White patients were included along with 144 non-White patients, comprising of Asian (16%), Black (26%), Hispanic (9%), and other/declined (50%) race. Statistical analysis was carried out using GraphPad Prism. RESULTS On multivariate analysis, White race was significantly associated with a later age at diagnosis independent of IDH1 status. White patients were more likely to present with a cognitive deficit (42.3% vs. 21.1%; p = 0.02*), while less likely to present with midline shift (32.5% vs. 49.3%; p = 0.004**) and mass effect on imaging (59.8% vs. 76.1%; p = 0.003***). Additionally, Black patients were more likely to present with syncope (15.8% vs. 2.3% [n = 107]; p = 0.04*) and Hispanic patients were more likely to present with seizure (35.7% vs. 15.9% [n = 110]; p = 0.03*). CONCLUSION White race appears to be independently associated with a later age at diagnosis of HGG. Furthermore, Black and Hispanic patients are more likely to present with severe, life-threatening symptoms. Large-scale studies are needed to elucidate race-based differences in HGG presentation to effectively predict outcomes.
Collapse
Affiliation(s)
- Sayak R Ghosh
- Department of Neurological Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, 3316 Rochambeau Ave, Bronx, NY, 10467, USA.
- Montefiore Medical Center, The University Hospital for the Albert Einstein College of Medicine, 3316 Rochambeau Avenue, Bronx, NY, 10467, USA.
| | - Anne R Lally
- Department of Neurological Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, 3316 Rochambeau Ave, Bronx, NY, 10467, USA
- Montefiore Medical Center, The University Hospital for the Albert Einstein College of Medicine, 3316 Rochambeau Avenue, Bronx, NY, 10467, USA
| | - Isabella L Pecorari
- Department of Neurological Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, 3316 Rochambeau Ave, Bronx, NY, 10467, USA
- Montefiore Medical Center, The University Hospital for the Albert Einstein College of Medicine, 3316 Rochambeau Avenue, Bronx, NY, 10467, USA
| | - Joshua Reynolds
- Department of Neurological Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, 3316 Rochambeau Ave, Bronx, NY, 10467, USA
- Montefiore Medical Center, The University Hospital for the Albert Einstein College of Medicine, 3316 Rochambeau Avenue, Bronx, NY, 10467, USA
| | - Alexander Ledet
- Department of Neurological Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, 3316 Rochambeau Ave, Bronx, NY, 10467, USA
- Montefiore Medical Center, The University Hospital for the Albert Einstein College of Medicine, 3316 Rochambeau Avenue, Bronx, NY, 10467, USA
| | - Sabrina Begley
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra Northwell, 500 Hofstra Blvd, Hempstead, NY, 11549, USA
| | - Elizabeth Juarez Diaz
- Department of Oncology, Washington University at St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Eric Zhu
- Department of Oncology, Washington University at St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Karan Joseph
- Department of Oncology, Washington University at St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Kyle McGeehan MPhil
- Department of Oncology, Washington University at St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Michael Schulder
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra Northwell, 500 Hofstra Blvd, Hempstead, NY, 11549, USA
| | - Tanner Johanns
- Department of Oncology, Washington University at St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110, USA
| | - Yonah C Ziemba
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra Northwell, 500 Hofstra Blvd, Hempstead, NY, 11549, USA
| | - Vijay Agarwal
- Department of Neurological Surgery, Albert Einstein College of Medicine/Montefiore Medical Center, 3316 Rochambeau Ave, Bronx, NY, 10467, USA
- Montefiore Medical Center, The University Hospital for the Albert Einstein College of Medicine, 3316 Rochambeau Avenue, Bronx, NY, 10467, USA
| |
Collapse
|
7
|
Hong WJ, Ho HW, Lin HM, Lin T, Chow WH, Yang CC, Lin LC. A Dosimetric Comparison of HyperArc Therapy Planning and Volumetric Modulated Arc Therapy Planning in Treating Patients With Glioblastoma Multiforme. In Vivo 2025; 39:1009-1021. [PMID: 40011001 PMCID: PMC11884473 DOI: 10.21873/invivo.13906] [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: 11/06/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 02/28/2025]
Abstract
BACKGROUND/AIM This study aimed at evaluating the potential benefit of automatic non-coplanar volumetric arc therapy (VMAT) (hyperarc, HA) technique in treating glioblastoma multiforme (GBM). PATIENTS AND METHODS Twenty-seven patients with GBM who received coplanar VMAT (C-VMAT) were selected in this study. HA and non-coplanar VMAT (NC-VMAT) plans were generated with the same prescriptions and constraints. The Target coverage, organs at risk (OARs) dose, and dosimetric indexes were compared among three plans. RESULTS The HA plan demonstrated a reduction in dose to normal tissues while maintaining target coverage, compared to C-VMAT and NC-VMAT. Additionally, HA plans demonstrated higher coverage of the GTV and PTV60 as well as improved CI from PTV60 and PTV46 compared to the other plans. Regarding the dose gradient, HA plans showed a greater dose fall-off, resulting in reduced high-dose and intermediate-dose spillage at PTV46 The HA also demonstrated a tighter gradient radius at PTV60 and PTV46 The HA plan requires fewer MUs than both C-VMAT and NC-VMAT. CONCLUSION The HA plan had better dosimetric results compared to C-VMAT and NC-VMAT. The HA with automatic planning module and auto-delivery treatment also provided high-quality planning and delivery efficacy. These advantages suggest that HA could potentially escalate tumor doses while minimizing toxicity, thereby improving outcomes in GBM patients.
Collapse
Affiliation(s)
- Wei-Ju Hong
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Hsiu-Wen Ho
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Hsiu-Man Lin
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Tung Lin
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Wan-Hsuan Chow
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan, R.O.C
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan, R.O.C.;
- Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan, R.O.C
| | - Li-Ching Lin
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan, R.O.C.;
| |
Collapse
|
8
|
Yu Y, Arrigo A, Chandra A, Zhuang C, Najjar MK, Khan MS, Zhu D, Dono A, Strowd RE, Tandon N, Zhu JJ, Hsu SH, Esquenazi Y, Chan M, Lo HW. Targeting tGLI1, a novel mediator of tumor therapeutic resistance, using Ketoconazole sensitizes glioblastoma to CDK4/6 therapy and chemoradiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639359. [PMID: 40060625 PMCID: PMC11888219 DOI: 10.1101/2025.02.20.639359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Glioblastoma (GBM) remains the most aggressive primary brain tumor in adults, with no effective treatments. While cyclin-dependent kinase 4/6 inhibitors (CDK4/6is) show clinical promise in some cancers, they have not significantly improved survival in GBM patients. This lack of response is attributed to the treatment-resistant glioma stem cell (GSC) population. We previously identified truncated glioma-associated oncogene homolog 1 (tGLI1) as a novel transcription factor promoting GSCs; however, its role in CDK4/6i resistance has never been investigated in any cancer type. Here, we found positive correlations between tGLI1 and CDK4/6 therapeutic resistance in patient datasets and in vitro studies. Pharmacological inhibition of tGLI1 using FDA-approved ketoconazole (KCZ), a tGLI1-specific inhibitor, sensitized GBM and GSCs to CDK4/6is. KCZ+CDK4/6i combination therapy demonstrated synergistic anti-proliferative effects, significantly inhibiting GBM stemness and cell cycle progression while increasing apoptosis. The combination was more efficacious than monotherapies in two orthotopic GBM mouse models. tGLI1 promoted GBM resistance to radiation therapy and temozolomide, while KCZ potentiated effects of these treatments. Collectively, we report for the first time that tGLI1 is a novel mediator of GBM resistance to CDK4/6is, and KCZ sensitizes GBM to CDK4/6is, thereby supporting future clinical utility of novel KCZ+CDK4/6i combinatorial therapy for GBM patients.
Collapse
Affiliation(s)
- Yang Yu
- Department of Cancer Biology, Wake Forest School University of Medicine, Winston-Salem, NC
| | - Austin Arrigo
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ankush Chandra
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chuling Zhuang
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mariana K Najjar
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Munazza S Khan
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dongqin Zhu
- Department of Cancer Biology, Wake Forest School University of Medicine, Winston-Salem, NC
| | - Antonio Dono
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Roy E Strowd
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Nitin Tandon
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jay-Jiguang Zhu
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sigmund H Hsu
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michael Chan
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Hui-Wen Lo
- Department of Cancer Biology, Wake Forest School University of Medicine, Winston-Salem, NC
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
9
|
Malik AA, Nguyen KC, Nardini JT, Krona CC, Flores KB, Nelander S. Mathematical modeling of multicellular tumor spheroids quantifies inter-patient and intra-tumor heterogeneity. NPJ Syst Biol Appl 2025; 11:20. [PMID: 39955270 PMCID: PMC11830081 DOI: 10.1038/s41540-025-00492-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 01/10/2025] [Indexed: 02/17/2025] Open
Abstract
In the study of brain tumors, patient-derived three-dimensional sphere cultures provide an important tool for studying emerging treatments. The growth of such spheroids depends on the combined effects of proliferation and migration of cells, but it is challenging to make accurate distinctions between increase in cell number versus the radial movement of cells. To address this, we formulate a novel model in the form of a system of two partial differential equations (PDEs) incorporating both migration and growth terms, and show that it more accurately fits our data compared to simpler PDE models. We show that traveling-wave speeds are strongly associated with population heterogeneity. Having fitted the model to our dataset we show that a subset of the cell lines are best described by a "Go-or-Grow"-type model, which constitutes a special case of our model. Finally, we investigate whether our fitted model parameters are correlated with patient age and survival.
Collapse
Affiliation(s)
- Adam A Malik
- Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden.
| | - Kyle C Nguyen
- Biomathematics Graduate Program, North Carolina State University, Raleigh, NC, USA
- Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC, USA
| | - John T Nardini
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA
| | - Cecilia C Krona
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kevin B Flores
- Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC, USA
- Department of Mathematics, North Carolina State University, Raleigh, NC, USA
| | - Sven Nelander
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Thanasupawat T, Mejia YP, Anandhan SS, Guo Y, Tiwana J, Fernando A, Glogowska A, Shafai T, daSilva S, Kaur N, Begum F, Zahedi R, Hombach-Klonisch S, Klonisch T. Proteomic and cytokine profiling of a CTRP8-RXFP1 glioma mouse model. Biochem Pharmacol 2025; 232:116722. [PMID: 39709036 DOI: 10.1016/j.bcp.2024.116722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Glioblastoma (GB) is the most prevalent and aggressive primary brain tumor with fatal outcome due to a lack of effective treatments. We previously identified C1q-tumor necrosis factor-related protein 8 (CTRP8), a new member of the adiponectin family, as a novel agonist of the relaxin family peptide receptor 1 (RXFP1) and showed that the CTRP8-RXFP1 ligand-receptor system facilitates increased invasiveness and chemoresistance in GB cells. In the present study, we have investigated the role of the CTRP8-RXFP1 signaling axis in glioma progression using an orthotopic mouse model xenografted with human U251 glioma cells stably expressing CTRP8 and RXFP1. Our results demonstrate that this in-vivo U251-CTRP8/RXFP1 glioma model promoted the formation of aggressive, highly proliferative glioma that resulted in significantly shorter survival times of xenografted mice. CTRP8/RXFP1 xenografts showed strongly elevated mitotic activity, increased expression of cathepsin B at the migrating front and promoted a pro-inflammatory tumor microenvironment characterized by a strong upregulation of cytokines, among them eotaxin-2 and-3, interleukin (IL)-6, IL-18 and others. Proteomic analysis of xenografted mouse brain identified both human and mouse proteome signatures unique to CTRP8/RXFP1 xenografts compared to U251 xenografts. In conclusion, our results suggest that co-expression of CTRP8 and RXFP1 promotes signaling pathways that generate unique tissue proteomic and inflammatory cytokine signatures which promote glioma aggressiveness. The CTRP-RXFP1 signaling pathway may represent an effective therapeutic target for the treatment of fast-progressing and currently untreatable GB.
Collapse
Affiliation(s)
| | | | | | - Yaxiong Guo
- Department of Human Anatomy and Cell Science, Winnipeg, MB, Canada; Department of Pathophysiology, Basic Medical College, Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Jasneet Tiwana
- Department of Human Anatomy and Cell Science, Winnipeg, MB, Canada
| | - Adline Fernando
- Department of Human Anatomy and Cell Science, Winnipeg, MB, Canada
| | | | - Talia Shafai
- Department of Human Anatomy and Cell Science, Winnipeg, MB, Canada
| | - Simone daSilva
- Department of Human Anatomy and Cell Science, Winnipeg, MB, Canada
| | - Nimrat Kaur
- Department of Human Anatomy and Cell Science, Winnipeg, MB, Canada
| | - Farhana Begum
- Department of Human Anatomy and Cell Science, Winnipeg, MB, Canada
| | - Rene Zahedi
- Manitoba Centre for Proteomics and Systems Biology (MCPSB), Winnipeg, MB, Canada
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, Winnipeg, MB, Canada; Department of Pathology, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, Winnipeg, MB, Canada; Department of Pathology, University of Manitoba, Rady Faculty of Health Sciences, Max Rady College of Medicine, Winnipeg, MB, Canada; CancerCare Manitoba, Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada.
| |
Collapse
|
11
|
Bumbaca B, Huggins JR, Birtwistle MR, Gallo JM. Network analyses of brain tumor multiomic data reveal pharmacological opportunities to alter cell state transitions. NPJ Syst Biol Appl 2025; 11:14. [PMID: 39893170 PMCID: PMC11787326 DOI: 10.1038/s41540-025-00493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
Glioblastoma Multiforme (GBM) remains a particularly difficult cancer to treat, and survival outcomes remain poor. In addition to the lack of dedicated drug discovery programs for GBM, extensive intratumor heterogeneity and epigenetic plasticity related to cell-state transitions are major roadblocks to successful drug therapy in GBM. To study these phenomenon, publicly available snRNAseq and bulk RNAseq data from patient samples were used to categorize cells from patients into four cell states (i.e., phenotypes), namely: (i) neural progenitor-like (NPC-like), (ii) oligodendrocyte progenitor-like (OPC-like), (iii) astrocyte-like (AC-like), and (iv) mesenchymal-like (MES-like). Patients were subsequently grouped into subpopulations based on which cell-state was the most dominant in their respective tumor. By incorporating phosphoproteomic measurements from the same patients, a protein-protein interaction network (PPIN) was constructed for each cell state. These four-cell state PPINs were pooled to form a single Boolean network that was used for in silico protein knockout simulations to investigate mechanisms that either promote or prevent cell state transitions. Simulation results were input into a boosted tree machine learning model which predicted the cell states or phenotypes of GBM patients from an independent public data source, the Glioma Longitudinal Analysis (GLASS) Consortium. Combining the simulation results and the machine learning predictions, we generated hypotheses for clinically relevant causal mechanisms of cell state transitions. For example, the transcription factor TFAP2A can be seen to promote a transition from the NPC-like to the MES-like state. Such protein nodes and the associated signaling pathways provide potential drug targets that can be further tested in vitro and support cell state-directed (CSD) therapy.
Collapse
Affiliation(s)
- Brandon Bumbaca
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Jonah R Huggins
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - James M Gallo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
12
|
Pandey R, Natarajan P, Reddy UK, Du W, Sirbu C, Sissoko M, Hankins GR. Deciphering the dose-dependent effects of thymoquinone on cellular proliferation and transcriptomic changes in A172 glioblastoma cells. PLoS One 2025; 20:e0318185. [PMID: 39874307 PMCID: PMC11774404 DOI: 10.1371/journal.pone.0318185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/11/2025] [Indexed: 01/30/2025] Open
Abstract
Glioblastoma multiforme (GBM), the most prevalent primary malignant brain tumor in adults, exhibits a dismal 6.9% five-year survival rate post-diagnosis. Thymoquinone (TQ), the most abundant bioactive compound in Nigella sativa, has been extensively researched for its anticancer properties across various human cancers. However, its specific anti-cancer mechanisms and pathways in glioblastoma remain to be completely elucidated. In this study, we assessed the impact of different TQ concentrations on the viability of A172 cells using WST-8 and Toluidine blue assays, followed by RNA sequencing (RNA-Seq) to identify differentially expressed genes (DEGs). We confirmed their expression levels through quantitative RT-PCR and performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses for these DEGs. RNA-seq revealed no significant gene expression changes at 2.5 μM and 5 μM TQ concentrations. However, at 25 μM and 50 μM, TQ significantly reduced cell viability dose-dependently. We identified 1548 DEGs at 25 μM TQ (684 up-regulated, 864 down-regulated) and 2797 DEGs at 50 μM TQ (1528 up-regulated, 1269 downregulated), with 1202 DEGs common to both concentrations. TQ inhibited key pathways such as PI3K-Akt signaling, calcium signaling, focal adhesion, and ECM-receptor interaction in A172 cells. It downregulated several potential oncogenes (e.g., AEBP1, MIAT) and genes linked to GBM proliferation and migration (e.g., SOCS2, HCP5) while modulating Wnt signaling and up-regulating tumor suppressor genes (e.g., SPRY4, BEX2). TQ also affected p53 downstream targets, maintaining p53 levels. This study elucidates the anti-cancer mechanisms of TQ in A172 GBM cells, underscoring its effects on multiple signaling pathways and positioning TQ as a promising candidate for innovative glioblastoma treatment strategies.
Collapse
Affiliation(s)
- Rachana Pandey
- Department of Biology, West Virginia State University, Institute, WV, United States of America
| | - Purushothaman Natarajan
- Department of Biology, West Virginia State University, Institute, WV, United States of America
- Department of Agriculture, Food, and Resource Sciences, University of Maryland Eastern Shore, Princess Anne, MD, United States of America
| | - Umesh K. Reddy
- Department of Biology, West Virginia State University, Institute, WV, United States of America
| | - Wei Du
- Cancer Center, Charleston Area Medical Center, Charleston, WV, United States of America
- Institute for Academic Medicine, Charleston, WV, United States of America
| | - Cristian Sirbu
- Cancer Center, Charleston Area Medical Center, Charleston, WV, United States of America
- Institute for Academic Medicine, Charleston, WV, United States of America
| | - Moussa Sissoko
- Katmai Oncology Group, Anchorage, Alaska, United States of America
| | - Gerald R. Hankins
- Department of Biology, West Virginia State University, Institute, WV, United States of America
| |
Collapse
|
13
|
Licón-Muñoz Y, Avalos V, Subramanian S, Granger B, Martinez F, García-Montaño LA, Varela S, Moore D, Perkins E, Kogan M, Berto S, Chohan MO, Bowers CA, Piccirillo SGM. Single-nucleus and spatial landscape of the sub-ventricular zone in human glioblastoma. Cell Rep 2025; 44:115149. [PMID: 39752252 DOI: 10.1016/j.celrep.2024.115149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/22/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The sub-ventricular zone (SVZ) is the most well-characterized neurogenic area in the mammalian brain. We previously showed that in 65% of patients with glioblastoma (GBM), the SVZ is a reservoir of cancer stem-like cells that contribute to treatment resistance and the emergence of recurrence. Here, we build a single-nucleus RNA-sequencing-based microenvironment landscape of the tumor mass and the SVZ of 15 patients and two histologically normal SVZ samples as controls. We identify a ZEB1-centered mesenchymal signature in the tumor cells of the SVZ. Moreover, the SVZ microenvironment is characterized by tumor-supportive microglia, which spatially coexist and establish crosstalks with tumor cells. Last, differential gene expression analyses, predictions of ligand-receptor and incoming/outgoing interactions, and functional assays reveal that the interleukin (IL)-1β/IL-1RAcP and Wnt-5a/Frizzled-3 pathways represent potential therapeutic targets in the SVZ. Our data provide insights into the biology of the SVZ in patients with GBM and identify potential targets of this microenvironment.
Collapse
Affiliation(s)
- Yamhilette Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Vanessa Avalos
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Suganya Subramanian
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bryan Granger
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Frank Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Leopoldo A García-Montaño
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Samantha Varela
- University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Drew Moore
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Eddie Perkins
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Michael Kogan
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM 87131, USA
| | - Stefano Berto
- Bioinformatics Core, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Neurogenomics Laboratory, Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Muhammad O Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Christian A Bowers
- Department of Neurosurgery, University of New Mexico Hospital, Albuquerque, NM 87131, USA
| | - Sara G M Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
14
|
Moses A, Malek R, Kendirli MT, Cheung P, Landry M, Herrera-Barrera M, Khojasteh A, Granucci M, Bukhari SA, Hooper JE, Hayden-Gephart M, Dixon SJ, Recht LD, Beinat C. Monitoring of cancer ferroptosis with [ 18F]hGTS13, a system xc- specific radiotracer. Theranostics 2025; 15:836-849. [PMID: 39776801 PMCID: PMC11700874 DOI: 10.7150/thno.101882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/16/2024] [Indexed: 01/11/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor in adults, characterized by resistance to conventional therapies and poor survival. Ferroptosis, a form of regulated cell death driven by lipid peroxidation, has recently emerged as a promising therapeutic target for GBM treatment. However, there are currently no non-invasive imaging techniques to monitor the engagement of pro-ferroptotic compounds with their respective targets, or to monitor the efficacy of ferroptosis-based therapies. System xc-, an important player in cellular redox homeostasis, plays a critical role in ferroptosis by mediating the exchange of cystine for glutamate, thus regulating the availability of cysteine, a crucial precursor for glutathione synthesis, and influencing the cellular antioxidant defense system. We have recently reported the development and validation of [18F]hGTS13, a radiopharmaceutical specific for system xc-. Methods: In the current work, we characterized the sensitivity of various cell lines to pro-ferroptotic compounds and evaluated the ability of [18F]hGTS13 to distinguish between sensitive and resistant cell lines and monitor changes in response to ferroptosis-inducing investigational compounds. We then associated changes in [18F]hGTS13 uptake with cellular glutathione content. Furthermore, we evaluated [18F]hGTS13 uptake in a rat model of glioma, both before and after treatment with imidazole ketone erastin (IKE), a pro-ferroptotic inhibitor of system xc- activity. Results: Treatment with erastin2, a system xc- inhibitor, significantly decreased [18F]hGTS13 uptake and cellular glutathione content in vitro. Dynamic PET/CT imaging of C6 glioma-bearing rats with [18F]hGTS13 revealed high and sustained uptake within the intracranial glioma and this uptake was decreased upon pre-treatment with IKE. Conclusion: In summary, [18F]hGTS13 represents a promising tool to distinguish cell types that demonstrate sensitivity or resistance to ferroptosis-inducing therapies that target system xc-, and monitor the engagement of these drugs.
Collapse
Affiliation(s)
- Abraham Moses
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Rim Malek
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Mustafa Tansel Kendirli
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Pierre Cheung
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Madeleine Landry
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Marco Herrera-Barrera
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Abbas Khojasteh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Monica Granucci
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Syed. A. Bukhari
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jody E. Hooper
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Melanie Hayden-Gephart
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Scott J. Dixon
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Lawrence D. Recht
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Corinne Beinat
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
15
|
Nawabi NLA, Saway BF, Jha R, Pereira M, Mehta NH, Das A, Zukas A, Lindhorst S, Strickland BA. Current trends in the allocation of National Institute of Health funding of brain tumor research. Neurooncol Adv 2025; 7:vdae203. [PMID: 40191402 PMCID: PMC11969036 DOI: 10.1093/noajnl/vdae203] [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] [Indexed: 04/09/2025] Open
Abstract
Background The National Institute of Health (NIH) provides a sizable annual budget toward brain tumor research. However, funding allocation for specific pathologies remains poorly described. We aimed to characterize the current landscape of NIH funding toward brain tumors as a function of pathology. Methods NIHRePORTER was queried to identify studies focused on glioblastoma, pediatric glioma, oligodendroglioma, brain metastasis, meningioma, pituitary adenoma, and vestibular schwannoma, from 2000 to 2023. Studies with R, U, and P funding mechanisms were included. Data were compiled and assessed according to pathology. Results Across these 7 tumors, 3320 unique studies with R, U, or P funding mechanisms were identified from 2000 to 2023. These were conducted across 480 unique institutions. The sum of funds allocated to all studies was $1 607 662 631. Glioblastoma commanded the largest portion of funds, representing 54% of R mechanisms, 55% of R01-funded studies, 48% of U mechanisms, and 49% of P mechanisms, and accounted for 51% ($813 556 423) of total funding. Brain metastasis was the second most-funded tumor, representing 31% of all R mechanisms, 31% of all R01-funded studies, 26% of all U mechanisms, and 28% of all P mechanisms, and accounted for 29% ($472 715 745) of funding. The remaining 14% of R mechanisms, 26% of U mechanisms, and 23% of P mechanisms focused on the remaining pathologies, and accounted for 20% ($321 390 463) of funding. Conclusions The current landscape of NIH funding for brain tumor research indicates that awarded mechanisms prioritize malignant intra-axial malignancies. Despite their prevalence, skull base neoplasia is far less represented in NIH-funded studies.
Collapse
Affiliation(s)
- Noah L A Nawabi
- College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Brian F Saway
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina
| | - Rohan Jha
- Harvard Medical School, Boston, Massachusetts
| | - Matheus Pereira
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina
| | | | - Arabinda Das
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina
| | - Alicia Zukas
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina
| | - Scott Lindhorst
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina
| | - Ben A Strickland
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
16
|
Zhong Z, Gan L, Feng Z, Wang W, Pan X, Wu C, Huang Y. Hydrogel local drug delivery systems for postsurgical management of tumors: Status Quo and perspectives. Mater Today Bio 2024; 29:101308. [PMID: 39525397 PMCID: PMC11550774 DOI: 10.1016/j.mtbio.2024.101308] [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: 08/06/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Surgery is one of the primary treatments for solid tumors. However, the incomplete resection of tumor cells and the immunosuppressive microenvironment make the issue of postsurgical tumor recurrence a great challenge. Furthermore, a wide range of requirements, including ensuring effective hemostasis, implementing prophylactic measures against infection, and promoting wound healing, were also raised in the postsurgical management of tumors. To fulfill these demands, multiple hydrogel local drug delivery systems (HLDDS) were developed recently. These HLDDS are expected to offer numerous advantages in the postsurgical management of tumors, such as achieving high local drug concentrations at the lesion, efficient delivery to surgical microcavities, mitigating systemic side effects, and addressing the diverse demand. Thus, in this review, a detailed discussion of the diverse demands of postsurgical management of tumors is provided. And the current publication trend on HLDDS in the postsurgical management of tumors is analyzed and discussed. Then, the applications of different types of HLDDS, in-situ HLDDS and non-in-situ HLDDS, in postsurgical management of tumors were introduced and summarized. Besides, the current problems and future perspectives are discussed. The review is expected to provide an overview of HLDDS in postsurgical management of tumors and promote their clinical application.
Collapse
Affiliation(s)
- Ziqiao Zhong
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| | - Lu Gan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| | - Ziyi Feng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| | - Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Chuanbin Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| | - Ying Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| |
Collapse
|
17
|
Rajkhowa S, Jha S. The role of NLRP3 and NLRP12 inflammasomes in glioblastoma. Genes Immun 2024; 25:541-551. [PMID: 39604503 DOI: 10.1038/s41435-024-00309-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Glioblastoma (GBM) is the deadliest malignant brain tumor, with a survival of less than 14 months after diagnosis. The highly invasive nature of GBM makes total surgical resection challenging, leading to tumor recurrence and declined survival. The heterocellular composition of the GBM reprograms its microenvironment, favoring tumor growth, proliferation, and migration. The innate immune cells in the GBM tumor microenvironment, including microglia, astrocytes, and macrophages, express pattern recognition receptors such as NLRs (Nucleotide-binding domain and leucine-rich repeat-containing) that sense pathogen- and damage-associated molecular patterns initiating inflammation. Upon activation, NLRP3 promotes inflammation by NLRP3 inflammasome formation. Auto-proteolytic cleavage and activation of Caspase-1 within the inflammasome leads to caspase-1-mediated cleavage, activation, and conversion of pro-IL-1ß and pro-IL-18 to IL-1ß and IL-18, leading to pyroptosis. In contrast, NLRP12 downregulates inflammatory responses in microglia and macrophages by regulating the NF-κB pathway. NLRP3 and NLRP12 have been implicated in the disease pathophysiology of several cancers with cell-context-dependent, pro- or anti-tumorigenic roles. In this review, we discuss the current literature on the mechanistic roles of NLRP3 and NLRP12 in GBM and the gaps in the scientific literature in the context of GBM pathophysiology with potential for targeted therapeutics.
Collapse
Affiliation(s)
- Sushmita Rajkhowa
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Sushmita Jha
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India.
| |
Collapse
|
18
|
Iorio AL, Lenci E, Marzano C, Bucaletti E, Tirinnanzi B, Casati G, Giunti L, Dallari C, Credi C, Sardi I, Trabocchi A. Oxime Linked Doxorubicin Glycoconjugates Improve the Specific Targeting of Glioblastoma in High-Grade Glioma Therapy. ACS Med Chem Lett 2024; 15:1953-1960. [PMID: 39563793 PMCID: PMC11571026 DOI: 10.1021/acsmedchemlett.4c00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
The treatment of glioblastoma (GBM) represents an urgent challenge for public health due to the inability to effectively deliver anticancer agents, such as doxorubicin (DOX), through the blood-brain barrier (BBB). Herein we report the synthesis of two novel DOX glycoconjugates using an oxime linkage that maintained the intercalation capability of the planar anthracycline ring of DOX, as demonstrated by UV-vis and fluorescence experiments in the presence of DNA. The biological effect of DOX glycoconjugates was evaluated in GBM cell lines, showing an enhanced cytotoxic and pro-apoptotic effect of 7 as compared to 4 and to conventional DOX. These data were confirmed in an in vitro coculture BBB model in which DOX glycoconjugate 7 showed high capability to cross a cellular monolayer and exert its cytotoxic effect on GBM cells. The results show that conjugation with glucose may represent a helpful tool to increase chemotherapy effectiveness in poor-responding GBM patients.
Collapse
Affiliation(s)
- Anna Lisa Iorio
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, 50139 Florence, Italy
| | - Elena Lenci
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 13, 50019 Sesto Fiorentino, Florence, Italy
| | - Chiara Marzano
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Elisabetta Bucaletti
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 13, 50019 Sesto Fiorentino, Florence, Italy
| | - Bianca Tirinnanzi
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Giacomo Casati
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Laura Giunti
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Caterina Dallari
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, 50019 Sesto Fiorentino, Florence, Italy
- National Institute of Optics National Research Council, 50019 Sesto Fiorentino, Florence, Italy
| | - Caterina Credi
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, 50019 Sesto Fiorentino, Florence, Italy
- National Institute of Optics National Research Council, 50019 Sesto Fiorentino, Florence, Italy
| | - Iacopo Sardi
- Neuro-Oncology Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
| | - Andrea Trabocchi
- Department of Chemistry "Ugo Schiff", University of Florence, via della Lastruccia 13, 50019 Sesto Fiorentino, Florence, Italy
| |
Collapse
|
19
|
Parès L, Naasri S, Delattre L, Therriault H, Liberelle B, De Crescenzo G, Lauzon MA, Faucheux N, Paquette B, Virgilio N. Macroporous chitosan/alginate hydrogels crosslinked with genipin accumulate and retain glioblastoma cancer cells. RSC Adv 2024; 14:35286-35304. [PMID: 39502176 PMCID: PMC11537210 DOI: 10.1039/d4ra06197g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Grade IV multiforme glioblastoma (GBM) is an aggressive cancer that remains incurable due to the GBM cells invading and proliferating in the surrounding healthy tissues, even after tumor resection. A new therapeutic paradigm to treat GBM is to attract and accumulate GBM cells in a macroporous hydrogel inserted in the surgical cavity after tumor resection, followed by a targeted high dose of radiotherapy. This work presents a molding-based method to prepare macroporous hydrogels composed of sodium alginate and chitosan, homogeneously mixed in solution using sodium bicarbonate, and subsequently crosslinked with genipin and calcium chloride. The gels display a blue color, the result of chitosan crosslinking with genipin, fully interconnected pores with an average diameter of 180 μm (and tunable over a wide range), with a compression modulus of 10 kPa, close to the value of brain tissues. The gels are stable in cell culture media and keep their integrity after radiation doses comparable to current GBM treatment levels. Finally, F98 GBM cells accumulate relatively homogeneously and are retained within the gels.
Collapse
Affiliation(s)
- Lauriane Parès
- Research Center for High Performance Polymer and Composite Systems (CREPEC), Department of Chemical Engineering, Polytechnique Montréal Montréal H3C 3A7 Québec Canada
| | - Sahar Naasri
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke Sherbrooke J1H 5N4 Québec Canada
| | - Lisa Delattre
- Research Center for High Performance Polymer and Composite Systems (CREPEC), Department of Chemical Engineering, Polytechnique Montréal Montréal H3C 3A7 Québec Canada
| | - Hélène Therriault
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke Sherbrooke J1H 5N4 Québec Canada
| | - Benoît Liberelle
- Department of Chemical Engineering, Polytechnique Montréal Montréal H3C 3A7 Québec Canada
| | - Gregory De Crescenzo
- Department of Chemical Engineering, Polytechnique Montréal Montréal H3C 3A7 Québec Canada
| | - Marc-Antoine Lauzon
- Department of Chemical and Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke Sherbrooke J1K 2R1 Québec Canada
| | - Nathalie Faucheux
- Department of Chemical and Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke Sherbrooke J1K 2R1 Québec Canada
| | - Benoit Paquette
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke Sherbrooke J1H 5N4 Québec Canada
| | - Nick Virgilio
- Research Center for High Performance Polymer and Composite Systems (CREPEC), Department of Chemical Engineering, Polytechnique Montréal Montréal H3C 3A7 Québec Canada
| |
Collapse
|
20
|
Dutt R, Thorpe C, Galileo DS. QSOX1 Modulates Glioblastoma Cell Proliferation and Migration In Vitro and Invasion In Vivo. Cancers (Basel) 2024; 16:3620. [PMID: 39518060 PMCID: PMC11545231 DOI: 10.3390/cancers16213620] [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: 09/14/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Quiescin Sulfhydryl Oxidase 1 (QSOX1) is an enzyme that catalyzes the oxidation of free thiols to generate disulfide bonds in a variety of proteins, including the cell surface and extracellular matrix. QSOX1 has been reported to be upregulated in a number of cancers, and the overexpression of QSOX1 has been correlated with aggressive cancers and poor patient prognosis. Glioblastoma (GBM) brain cancer has been practically impossible to treat effectively, with cells that rapidly invade normal brain tissue and escape surgery and other treatment. Thus, there is a crucial need to understand the multiple mechanisms that facilitate GBM cell invasion and to determine if QSOX1 is involved. Methods and Results: Here, we investigated the function of QSOX1 in human glioblastoma cells using two cell lines derived from T98G cells, whose proliferation, motility, and invasiveness has been shown by us to be dependent on disulfide bond-containing adhesion and receptor proteins, such as L1CAM and the FGFR. We lentivirally introduced shRNA to attenuate the QSOX1 protein expression in one cell line, and a Western blot analysis confirmed the decreased QSOX1 expression. A DNA content/cell cycle analysis using flow cytometry revealed 27% fewer knockdown cells in the S-phase of the cell cycle, indicating a reduced proliferation. A cell motility analysis utilizing our highly quantitative SuperScratch time-lapse microscopy assay revealed that knockdown cells migrated more slowly, with a 45% decrease in migration velocity. Motility was partly rescued by the co-culture of knockdown cells with control cells, indicating a paracrine effect. Surprisingly, knockdown cells exhibited increased motility when assayed using a Transwell migration assay. Our novel chick embryo orthotopic xenograft model was used to assess the in vivo invasiveness of knockdown vs. control cells, and tumors developed from both cell types. However, fewer invasive knockdown cells were observed after about a week. Conclusions: Our results indicate that an experimental reduction in QSOX1 expression in GBM cells leads to decreased cell proliferation, altered in vitro migration, and decreased in vivo invasion.
Collapse
Affiliation(s)
- Reetika Dutt
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA; (R.D.); (C.T.)
| | - Colin Thorpe
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA; (R.D.); (C.T.)
| | - Deni S. Galileo
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
21
|
Rajesh R U, Sangeetha D. Therapeutic potentials and targeting strategies of quercetin on cancer cells: Challenges and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155902. [PMID: 39059266 DOI: 10.1016/j.phymed.2024.155902] [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: 03/07/2024] [Revised: 07/08/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Every cell in the human body is vital because it maintains equilibrium and carries out a variety of tasks, including growth and development. These activities are carried out by a set of instructions carried by many different genes and organized into DNA. It is well recognized that some lifestyle decisions, like using tobacco, alcohol, UV, or multiple sexual partners, might increase one's risk of developing cancer. The advantages of natural products for any health issue are well known, and researchers are making attempts to separate flavonoid-containing substances from plants. Various parts of plants contain a phenolic compound called flavonoid. Quercetin, which belongs to the class of compounds known as flavones with chromone skeletal structure, has anti-cancer activity. PURPOSE The study was aimed at investigating the therapeutic action of the flavonoid quercetin on various cancer cells. METHODS The phrases quercetin, anti-cancer, nanoparticles, and cell line were used to search the data using online resources such as PubMed, and Google Scholar. Several critical previous studies have been included. RESULTS Quercetin inhibits various dysregulated signaling pathways that cause cancer cells to undergo apoptosis to exercise its anticancer effects. Numerous signaling pathways are impacted by quercetin, such as the Hedgehog system, Akt, NF-κB pathway, downregulated mutant p53, JAK/STAT, G1 phase arrest, Wnt/β-Catenin, and MAPK. There are downsides to quercetin, like hydrophobicity, first-pass effect, instability in the gastrointestinal tract, etc., because of which it is not well-established in the pharmaceutical industry. The solution to these drawbacks in the future is using bio-nanomaterials like chitosan, PLGA, liposomes, and silk fibroin as carriers, which can enhance the target specificity of quercetin. The first section of this review covers the specifics of flavonoids and quercetin; the second section covers the anti-cancer activity of quercetin; and the third section explains the drawbacks and conjugation of quercetin with nanoparticles for drug delivery by overcoming quercetin's drawback. CONCLUSIONS Overall, this review presented details about quercetin, which is a plant derivative with a promising molecular mechanism of action. They inhibit cancer by various mechanisms with little or no side effects. It is anticipated that plant-based materials will become increasingly relevant in the treatment of cancer.
Collapse
Affiliation(s)
- Udaya Rajesh R
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India
| | - Dhanaraj Sangeetha
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India.
| |
Collapse
|
22
|
Sterner RC, Sterner RM. EGFRVIII and EGFR targeted chimeric antigen receptor T cell therapy in glioblastoma. Front Oncol 2024; 14:1434495. [PMID: 39364321 PMCID: PMC11446898 DOI: 10.3389/fonc.2024.1434495] [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: 05/17/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024] Open
Abstract
Glioblastoma is the most common primary brain tumor. Although there have been significant advances in surgical techniques, chemo and immunotherapies, and radiation therapy, outcomes continue to be devastating for these patients with minimal improvements in survival. Chimeric antigen receptor T cell therapy is a revolutionary approach that is a new pillar in the treatment of cancer. CAR T cell therapy has produced remarkable results in hematological malignancies; however, multiple limitations currently prevent it from being a first-line therapy, especially for solid tumors. Epidermal growth factor receptor is classically amplified in glioblastoma, and a variant, EGFR variant III, is expressed on glioblastoma, making it an exciting potential target for CAR T cell therapy. Although preclinical has exciting potential, clinical data has been heterogeneous. In this review, we assess the state of field of EGFR-targeted CAR T cells.
Collapse
Affiliation(s)
- Robert C Sterner
- Department of Neurosurgery, Inova Fairfax Medical Campus, Fairfax, VA, United States
| | - Rosalie M Sterner
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
23
|
Wei J, Wang M, Li S, Han R, Xu W, Zhao A, Yu Q, Li H, Li M, Chi G. Reprogramming of astrocytes and glioma cells into neurons for central nervous system repair and glioblastoma therapy. Biomed Pharmacother 2024; 176:116806. [PMID: 38796971 DOI: 10.1016/j.biopha.2024.116806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Central nervous system (CNS) damage is usually irreversible owing to the limited regenerative capability of neurons. Following CNS injury, astrocytes are reactively activated and are the key cells involved in post-injury repair mechanisms. Consequently, research on the reprogramming of reactive astrocytes into neurons could provide new directions for the restoration of neural function after CNS injury and in the promotion of recovery in various neurodegenerative diseases. This review aims to provide an overview of the means through which reactive astrocytes around lesions can be reprogrammed into neurons, to elucidate the intrinsic connection between the two cell types from a neurogenesis perspective, and to summarize what is known about the neurotranscription factors, small-molecule compounds and MicroRNA that play major roles in astrocyte reprogramming. As the malignant proliferation of astrocytes promotes the development of glioblastoma multiforme (GBM), this review also examines the research advances on and the theoretical basis for the reprogramming of GBM cells into neurons and discusses the advantages of such approaches over traditional treatment modalities. This comprehensive review provides new insights into the field of GBM therapy and theoretical insights into the mechanisms of neurological recovery following neurological injury and in GBM treatment.
Collapse
Affiliation(s)
- Junyuan Wei
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Shilin Li
- School of Public Health, Jilin University, Changchun 130021, China.
| | - Rui Han
- Department of Neurovascular Surgery, First Hospital of Jilin University, 1xinmin Avenue, Changchun, Jilin Province 130021, China.
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Anqi Zhao
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Qi Yu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Haokun Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
24
|
Bumbaca B, Birtwistle MR, Gallo JM. Network Analyses of Brain Tumor Patients' Multiomic Data Reveals Pharmacological Opportunities to Alter Cell State Transitions. RESEARCH SQUARE 2024:rs.3.rs-4391296. [PMID: 38826227 PMCID: PMC11142360 DOI: 10.21203/rs.3.rs-4391296/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Glioblastoma Multiforme (GBM) remains a particularly difficult cancer to treat, and survival outcomes remain poor. In addition to the lack of dedicated drug discovery programs for GBM, extensive intratumor heterogeneity and epigenetic plasticity related to cell-state transitions are major roadblocks to successful drug therapy in GBM. To study these phenomenon, publicly available snRNAseq and bulk RNAseq data from patient samples were used to categorize cells from patients into four cell states (i.e. phenotypes), namely: (i) neural progenitor-like (NPC-like), (ii) oligodendrocyte progenitor-like (OPC-like), (iii) astrocyte- like (AC-like), and (iv) mesenchymal-like (MES-like). Patients were subsequently grouped into subpopulations based on which cell-state was the most dominant in their respective tumor. By incorporating phosphoproteomic measurements from the same patients, a protein-protein interaction network (PPIN) was constructed for each cell state. These four-cell state PPINs were pooled to form a single Boolean network that was used for in silico protein knockout simulations to investigate mechanisms that either promote or prevent cell state transitions. Simulation results were input into a boosted tree machine learning model which predicted the cell states or phenotypes of GBM patients from an independent public data source, the Glioma Longitudinal Analysis (GLASS) Consortium. Combining the simulation results and the machine learning predictions, we generated hypotheses for clinically relevant causal mechanisms of cell state transitions. For example, the transcription factor TFAP2A can be seen to promote a transition from the NPC-like to the MES-like state. Such protein nodes and the associated signaling pathways provide potential drug targets that can be further tested in vitro and support cell state-directed (CSD) therapy.
Collapse
Affiliation(s)
- Brandon Bumbaca
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo NY, USA
| | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson SC, USA
- Department of Bioengineering, Clemson University, Clemson SC, USA
| | - James M Gallo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo NY, USA
| |
Collapse
|
25
|
Williams TL, Nwokoye P, Kuc RE, Smith K, Paterson AL, Allinson K, Maguire JJ, Davenport AP. Expression of the apelin receptor, a novel potential therapeutic target, and its endogenous ligands in diverse stem cell populations in human glioblastoma. Front Neurosci 2024; 18:1379658. [PMID: 38803685 PMCID: PMC11128631 DOI: 10.3389/fnins.2024.1379658] [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: 01/31/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and lethal forms of brain cancer, carrying a very poor prognosis (median survival of ~15 months post-diagnosis). Treatment typically involves invasive surgical resection of the tumour mass, followed by radiotherapy and adjuvant chemotherapy using the alkylating agent temozolomide, but over half of patients do not respond to this drug and considerable resistance is observed. Tumour heterogeneity is the main cause of therapeutic failure, where diverse progenitor glioblastoma stem cell (GSC) lineages in the microenvironment drive tumour recurrence and therapeutic resistance. The apelin receptor is a class A GPCR that binds two endogenous peptide ligands, apelin and ELA, and plays a role in the proliferation and survival of cancer cells. Here, we used quantitative whole slide immunofluorescent imaging of human GBM samples to characterise expression of the apelin receptor and both its ligands in the distinct GSC lineages, namely neural-progenitor-like cells (NPCs), oligodendrocyte-progenitor-like cells (OPCs), and mesenchymal-like cells (MES), as well as reactive astrocytic cells. The data confirm the presence of the apelin receptor as a tractable drug target that is common across the key cell populations driving tumour growth and maintenance, offering a potential novel therapeutic approach for patients with GBM.
Collapse
Affiliation(s)
- Thomas L. Williams
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Peter Nwokoye
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Rhoda E. Kuc
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Kieran Smith
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Anna L. Paterson
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Kieren Allinson
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Janet J. Maguire
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Anthony P. Davenport
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| |
Collapse
|
26
|
Bumbaca B, Birtwistle MR, Gallo JM. Network Analyses of Brain Tumor Patients' Multiomic Data Reveals Pharmacological Opportunities to Alter Cell State Transitions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593202. [PMID: 38766170 PMCID: PMC11100715 DOI: 10.1101/2024.05.08.593202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Glioblastoma Multiforme (GBM) remains a particularly difficult cancer to treat, and survival outcomes remain poor. In addition to the lack of dedicated drug discovery programs for GBM, extensive intratumor heterogeneity and epigenetic plasticity related to cell-state transitions are major roadblocks to successful drug therapy in GBM. To study these phenomenon, publicly available snRNAseq and bulk RNAseq data from patient samples were used to categorize cells from patients into four cell states (i.e. phenotypes), namely: (i) neural progenitor-like (NPC-like), (ii) oligodendrocyte progenitor-like (OPC-like), (iii) astrocyte-like (AC-like), and (iv) mesenchymal-like (MES-like). Patients were subsequently grouped into subpopulations based on which cell-state was the most dominant in their respective tumor. By incorporating phosphoproteomic measurements from the same patients, a protein-protein interaction network (PPIN) was constructed for each cell state. These four-cell state PPINs were pooled to form a single Boolean network that was used for in silico protein knockout simulations to investigate mechanisms that either promote or prevent cell state transitions. Simulation results were input into a boosted tree machine learning model which predicted the cell states or phenotypes of GBM patients from an independent public data source, the Glioma Longitudinal Analysis (GLASS) Consortium. Combining the simulation results and the machine learning predictions, we generated hypotheses for clinically relevant causal mechanisms of cell state transitions. For example, the transcription factor TFAP2A can be seen to promote a transition from the NPC-like to the MES-like state. Such protein nodes and the associated signaling pathways provide potential drug targets that can be further tested in vitro and support cell state-directed (CSD) therapy.
Collapse
Affiliation(s)
- Brandon Bumbaca
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo NY, USA
| | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson SC, USA
- Department of Bioengineering, Clemson University, Clemson SC, USA
| | - James M Gallo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo NY, USA
| |
Collapse
|
27
|
Shetty K, Yadav KS. Temozolomide nano-in-nanofiber delivery system with sustained release and enhanced cellular uptake by U87MG cells. Drug Dev Ind Pharm 2024; 50:420-431. [PMID: 38502031 DOI: 10.1080/03639045.2024.2332906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVE The study was aimed at formulating temozolomide (TMZ) loaded gelatin nanoparticles (GNPs) encapsulated into polyvinyl alcohol (PVA) nanofibers (TMZ-GNPs-PVA NFs) as the nano-in-nanofiber delivery system. The secondary objective was to explore the sustained releasing ability of this system and to assess its enhanced cellular uptake against U87MG glioma cells in vitro. SIGNIFICANCE Nano-in-nanofibers are the emerging drug delivery systems for treating a wide range of diseases including cancers as they overcome the challenges experienced by nanoparticles and nanofibers alone. METHODS The drug-loaded GNPs were formulated by one-step desolvation method. The Design of Experiments (DoE) was used to optimize nanoparticle size and entrapment efficiency. The optimized drug-loaded nanoparticles were then encapsulated within nanofibers using blend electrospinning technique. The U87MG glioma cells were used to investigate the uptake of the formulation. RESULTS A 32 factorial design was used to optimize the mean particle size (145.7 nm) and entrapment efficiency (87.6%) of the TMZ-loaded GNPs which were subsequently ingrained into PVA nanofibers by electrospinning technique. The delivery system achieved a sustained drug release for up to seven days (in vitro). The SEM results ensured that the expected nano-in-nanofiber delivery system was achieved. The uptake of TMZ-GNPs-PVA NFs by cells was increased by a factor of 1.964 compared to that of the pure drug. CONCLUSION The nano-in-nanofiber drug delivery system is a potentially useful therapeutic strategy for the management of glioblastoma multiforme.
Collapse
Affiliation(s)
- Karishma Shetty
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS (Deemed to be University), Mumbai, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS (Deemed to be University), Mumbai, India
| |
Collapse
|
28
|
Horváth L, Biri-Kovács B, Baranyai Z, Stipsicz B, Méhes E, Jezsó B, Krátký M, Vinšová J, Bősze S. New Salicylanilide Derivatives and Their Peptide Conjugates as Anticancer Compounds: Synthesis, Characterization, and In Vitro Effect on Glioblastoma. ACS OMEGA 2024; 9:16927-16948. [PMID: 38645331 PMCID: PMC11024950 DOI: 10.1021/acsomega.3c05727] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 04/23/2024]
Abstract
Pharmacologically active salicylanilides (2-hydroxy-N-phenylbenzamides) have been a promising area of interest in medicinal chemistry-related research for quite some time. This group of compounds has shown a wide spectrum of biological activities, including but not limited to anticancer effects. In this study, substituted salicylanilides were chosen to evaluate the in vitro activity on U87 human glioblastoma (GBM) cells. The parent salicylanilide, salicylanilide 5-chloropyrazinoates, a 4-aminosalicylic acid derivative, and the new salicylanilide 4-formylbenzoates were chemically and in vitro characterized. To enhance the internalization of the compounds, they were conjugated to delivery peptides with the formation of oxime bonds. Oligotuftsins ([TKPKG]n, n = 1-4), the ligands of neuropilin receptors, were used as GBM-targeting carrier peptides. The in vitro cellular uptake, intracellular localization, and penetration ability on tissue-mimicking models of the fluorescent peptide derivatives were determined. The compounds and their peptide conjugates significantly decreased the viability of U87 glioma cells. Salicylanilide compound-induced GBM cell death was associated with activation of autophagy, as characterized by immunodetection of autophagy-related processing of light chain 3 protein.
Collapse
Affiliation(s)
- Lilla Horváth
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Beáta Biri-Kovács
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Zsuzsa Baranyai
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Bence Stipsicz
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
- Institute
of Biology, Doctoral School of Biology, Eötvös Loránd University, Budapest 1117, Hungary
| | - Előd Méhes
- Institute
of Physics, Department of Biological Physics, Eötvös Loránd University, Budapest 1117, Hungary
| | - Bálint Jezsó
- Research
Centre for Natural Sciences, Institute of
Enzymology, Budapest 1053, Hungary
- ELTE-MTA
“Momentum” Motor Enzymology Research Group, Department
of Biochemistry, Eötvös Loránd
University, Budapest 1117, Hungary
| | - Martin Krátký
- Department
of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec
Králové, Charles University, 500 03 Hradec Králové, Czech Republic
| | - Jarmila Vinšová
- Department
of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec
Králové, Charles University, 500 03 Hradec Králové, Czech Republic
| | - Szilvia Bősze
- ELKH-ELTE
Research Group of Peptide Chemistry, Eötvös Loránd
Research Network, Eötvös Loránd
University, Budapest 1117, Hungary
| |
Collapse
|
29
|
Soleiman M, Fathi-Roudsari M, Khajeh K, Maghsoudi A. Optimization of Epigenetic Modifier Drug Combination for Synergistic Effect against Glioblastoma Multiform Cancer Cell Lines. Cancer Invest 2024; 42:319-332. [PMID: 38695671 DOI: 10.1080/07357907.2024.2345183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/16/2024] [Indexed: 05/28/2024]
Abstract
Glioblastoma multiforme (GBM), is a frequent class of malignant brain tumors. Epigenetic therapy, especially with synergistic combinations is highly paid attention for aggressive solid tumors like GBM. Here, RSM optimization has been used to increase the efficient arrest of U87 and U251 cell lines due to synergistic effects. Cell lines were treated with SAHA, 5-Azacytidine, GSK-126, and PTC-209 individually and then RSM was used to find most effective combinations. Results showed that optimized combinations significantly reduce cell survival and induce cell cycle arrest and apoptosis in both cell lines. Expression of cyclin B1 and cyclin D1 were decreased while caspase3 increased expression.
Collapse
Affiliation(s)
- Morvarid Soleiman
- Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mehrnoosh Fathi-Roudsari
- Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Khosro Khajeh
- Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
30
|
Sharma S, Wang SA, Yang WB, Lin HY, Lai MJ, Chen HC, Kao TY, Hsu FL, Nepali K, Hsu TI, Liou JP. First-in-Class Dual EZH2-HSP90 Inhibitor Eliciting Striking Antiglioblastoma Activity In Vitro and In Vivo. J Med Chem 2024; 67:2963-2985. [PMID: 38285511 PMCID: PMC10895674 DOI: 10.1021/acs.jmedchem.3c02053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
Structural analysis of tazemetostat, an FDA-approved EZH2 inhibitor, led us to pinpoint a suitable site for appendage with a pharmacophoric fragment of second-generation HSP90 inhibitors. Resultantly, a magnificent dual EZH2/HSP90 inhibitor was pinpointed that exerted striking cell growth inhibitory efficacy against TMZ-resistant Glioblastoma (GBM) cell lines. Exhaustive explorations of chemical probe 7 led to several revelations such as (i) compound 7 increased apoptosis/necrosis-related gene expression, whereas decreased M phase/kinetochore/spindle-related gene expression as well as CENPs protein expression in Pt3R cells; (ii) dual inhibitor 7 induced cell cycle arrest at the M phase; (iii) compound 7 suppressed reactive oxygen species (ROS) catabolism pathway, causing the death of TMZ-resistant GBM cells; and (iv) compound 7 elicited substantial in vivo anti-GBM efficacy in experimental mice xenografted with TMZ-resistant Pt3R cells. Collectively, the study results confirm the potential of dual EZH2-HSP90 inhibitor 7 as a tractable anti-GBM agent.
Collapse
Affiliation(s)
- Sachin Sharma
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
| | - Shao-An Wang
- School
of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Bin Yang
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
| | - Hong-Yi Lin
- Graduate
Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Mei-Jung Lai
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
| | - Hsien-Chung Chen
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
- Department
of Neurosurgery, Shuang Ho Hospital, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
| | - Tzu-Yuan Kao
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
| | - Feng-Lin Hsu
- School
of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Kunal Nepali
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-I Hsu
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
- International
Master Program in Medical Neuroscience, College of Medical Science
and Technology, Taipei Medical University, Taipei 110, Taiwan
- TMU
Research Center of Cancer Translational Medicine, Taipei 110 Taiwan
| | - Jing-Ping Liou
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center of Cancer Translational Medicine, Taipei 110 Taiwan
- Ph.D.
Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
31
|
Abdel-Rahman SA, Gabr M. Small Molecule Immunomodulators as Next-Generation Therapeutics for Glioblastoma. Cancers (Basel) 2024; 16:435. [PMID: 38275876 PMCID: PMC10814352 DOI: 10.3390/cancers16020435] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Glioblastoma (GBM), the most aggressive astrocytic glioma, remains a therapeutic challenge despite multimodal approaches. Immunotherapy holds promise, but its efficacy is hindered by the highly immunosuppressive GBM microenvironment. This review underscores the urgent need to comprehend the intricate interactions between glioma and immune cells, shaping the immunosuppressive tumor microenvironment (TME) in GBM. Immunotherapeutic advancements have shown limited success, prompting exploration of immunomodulatory approaches targeting tumor-associated macrophages (TAMs) and microglia, constituting a substantial portion of the GBM TME. Converting protumor M2-like TAMs to antitumor M1-like phenotypes emerges as a potential therapeutic strategy for GBM. The blood-brain barrier (BBB) poses an additional challenge to successful immunotherapy, restricting drug delivery to GBM TME. Research efforts to enhance BBB permeability have mainly focused on small molecules, which can traverse the BBB more effectively than biologics. Despite over 200 clinical trials for GBM, studies on small molecule immunomodulators within the GBM TME are scarce. Developing small molecules with optimal brain penetration and selectivity against immunomodulatory pathways presents a promising avenue for combination therapies in GBM. This comprehensive review discusses various immunomodulatory pathways in GBM progression with a focus on immune checkpoints and TAM-related targets. The exploration of such molecules, with the capacity to selectively target key immunomodulatory pathways and penetrate the BBB, holds the key to unlocking new combination therapy approaches for GBM.
Collapse
Affiliation(s)
- Somaya A. Abdel-Rahman
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Moustafa Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
32
|
Darwish A, Pammer M, Gallyas F, Vígh L, Balogi Z, Juhász K. Emerging Lipid Targets in Glioblastoma. Cancers (Basel) 2024; 16:397. [PMID: 38254886 PMCID: PMC10814456 DOI: 10.3390/cancers16020397] [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: 12/14/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
GBM accounts for most of the fatal brain cancer cases, making it one of the deadliest tumor types. GBM is characterized by severe progression and poor prognosis with a short survival upon conventional chemo- and radiotherapy. In order to improve therapeutic efficiency, considerable efforts have been made to target various features of GBM. One of the targetable features of GBM is the rewired lipid metabolism that contributes to the tumor's aggressive growth and penetration into the surrounding brain tissue. Lipid reprogramming allows GBM to acquire survival, proliferation, and invasion benefits as well as supportive modulation of the tumor microenvironment. Several attempts have been made to find novel therapeutic approaches by exploiting the lipid metabolic reprogramming in GBM. In recent studies, various components of de novo lipogenesis, fatty acid oxidation, lipid uptake, and prostaglandin synthesis have been considered promising targets in GBM. Emerging data also suggest a significant role hence therapeutic potential of the endocannabinoid metabolic pathway in GBM. Here we review the lipid-related GBM characteristics in detail and highlight specific targets with their potential therapeutic use in novel antitumor approaches.
Collapse
Affiliation(s)
- Ammar Darwish
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Milán Pammer
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Ferenc Gallyas
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - László Vígh
- Institute of Biochemistry, HUN-REN Biological Research Center, 6726 Szeged, Hungary
| | - Zsolt Balogi
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Kata Juhász
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
33
|
Repossi R, Martín-Ramírez R, Gómez-Bernal F, Medina L, Fariña-Jerónimo H, González-Fernández R, Martín-Vasallo P, Plata-Bello J. Evaluation of Zonulin Expression and Its Potential Clinical Significance in Glioblastoma. Cancers (Basel) 2024; 16:356. [PMID: 38254845 PMCID: PMC10814510 DOI: 10.3390/cancers16020356] [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: 12/08/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Glioblastoma, the deadliest adult brain tumor, poses a significant therapeutic challenge with a dismal prognosis despite current treatments. Zonulin, a protein influencing tight junctions and barrier functions, has gained attention for its diverse roles in various diseases. This study aimed to preliminarily analyze the circulating and tumor zonulin levels, evaluating their impact on disease prognosis and clinical-radiological factors. Additionally, we investigated in vitro zonulin expression in different glioblastoma cell lines under two different conditions. The study comprised 34 newly diagnosed glioblastoma patients, with blood samples collected before treatment for zonulin and haptoglobin analysis. Tumor tissue samples from 21 patients were obtained for zonulin expression. Clinical, molecular, and radiological data were collected, and zonulin protein levels were assessed using ELISA and Western blot techniques. Furthermore, zonulin expression was analyzed in vitro in three glioblastoma cell lines cultured under standard and glioma-stem-cell (GSC)-specific conditions. High zonulin expression in glioblastoma tumors correlated with larger preoperative contrast enhancement and edema volumes. Patients with high zonulin levels showed a poorer prognosis (progression-free survival [PFS]). Similarly, elevated serum levels of zonulin were associated with a trend of shorter PFS. Higher haptoglobin levels correlated with MGMT methylation and longer PFS. In vitro, glioblastoma cell lines expressed zonulin under standard cell culture conditions, with increased expression in tumorsphere-specific conditions. Elevated zonulin levels in both the tumor and serum of glioblastoma patients were linked to a poorer prognosis and radiological signs of increased disruption of the blood-brain barrier. In vitro, zonulin expression exhibited a significant increase in tumorspheres.
Collapse
Affiliation(s)
- Roberta Repossi
- Neurogenetics of Rare Disease Group, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Clinical Neuroscience Research Group, University of La Laguna, 38320 La Laguna, Spain
| | - Rita Martín-Ramírez
- Clinical Neuroscience Research Group, University of La Laguna, 38320 La Laguna, Spain
- Department of Molecular Biology, Faculty of Biology, University of La Laguna, 38320 La Laguna, Spain
| | - Fuensanta Gómez-Bernal
- Department of Biochemistry, Hospital Universitario de Canarias, 38320 S/C de Tenerife, Spain
| | - Lilian Medina
- Department of Biochemistry, Hospital Universitario de Canarias, 38320 S/C de Tenerife, Spain
| | - Helga Fariña-Jerónimo
- Clinical Neuroscience Research Group, University of La Laguna, 38320 La Laguna, Spain
- Department of Neurosurgery, Hospital Universitario de Canarias, 38320 S/C de Tenerife, Spain
| | - Rebeca González-Fernández
- Department of Molecular Biology, Faculty of Biology, University of La Laguna, 38320 La Laguna, Spain
| | - Pablo Martín-Vasallo
- Department of Molecular Biology, Faculty of Biology, University of La Laguna, 38320 La Laguna, Spain
| | - Julio Plata-Bello
- Clinical Neuroscience Research Group, University of La Laguna, 38320 La Laguna, Spain
- Department of Neurosurgery, Hospital Universitario de Canarias, 38320 S/C de Tenerife, Spain
| |
Collapse
|
34
|
Vazifehmand R, Ali DS, Homaie FM, Jalalvand FM, Othman Z, Deming C, Stanslas J, Sekawi Z. Effects of HSV-G47Δ Oncolytic Virus on Telomerase and Telomere Length Alterations in Glioblastoma Multiforme Cancer Stem Cells Under Hypoxia and Normoxia Conditions. Curr Cancer Drug Targets 2024; 24:1262-1274. [PMID: 38357955 DOI: 10.2174/0115680096274769240115165344] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/07/2023] [Accepted: 01/01/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Due to the existence of tumor stem cells with tumorigenicity properties and resistance patterns, treatment of glioblastoma is not easy. Hypoxia is a major concern in glioblastoma therapy. Telomerase activity and telomere length alterations have been known to play a critical role in glioblastoma progression and invasion. OBJECTIVE This study aimed to investigate the effects of HSV-G47Δ oncolytic virus on telomerase and telomere length alterations in U251GBMCSCs (U251-Glioblastoma cancer stem cells) under hypoxia and normoxia conditions. METHODS U251-CSCs were exposed to the HSV-G47Δ virus in optimized MOI (Multiplicity of infection= 1/14 hours). An absolute telomere length and gene expression of telomerase subunits were determined using an absolute human telomere length quantification PCR assay. Furthermore, a bioinformatics pathway analysis was carried out to evaluate physical and genetic interactions between dysregulated genes with other potential genes and pathways. RESULTS Data revealed that U251CSCs had longer telomeres when exposed to HSV-G47Δ in normoxic conditions but had significantly shorter telomeres in hypoxic conditions. Furthermore, hTERC, DKC1, and TEP1 genes were significantly dysregulated in hypoxic and normoxic microenvironments. The analysis revealed that the expression of TERF2 was significantly reduced in both microenvironments, and two critical genes from the MRN complex, MER11 and RAD50, were significantly upregulated in normoxic conditions. RAD50 showed a significant downregulation pattern in the hypoxic niche. Our results suggested that repair complex in the telomeric structure could be targeted by HSV-G47Δ in both microenvironments. CONCLUSION In the glioblastoma treatment strategy, telomerase and telomere complex could be potential targets for HSV-G47Δ in both microenvironments.
Collapse
Affiliation(s)
- Reza Vazifehmand
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Dhuha Saeed Ali
- Halal Products Research Institute, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | | | | | - Zulkefley Othman
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Chau Deming
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | - Zamberi Sekawi
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), 43400, Serdang, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
35
|
Joseph C. Guess What Is in My Brain. J Adv Pract Oncol 2024; 15:60-64. [PMID: 39119082 PMCID: PMC11308535 DOI: 10.6004/jadpro.2024.15.1.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
Magnetic resonance imaging (MRI) of the brain is an important diagnostic tool used by neurologists. This article explores the workup and management for a patient with a brain lesion and highlights the importance of neuroimaging. Similarities and differences in MRI findings for meningioma, central nervous system lymphoma, and glioblastomas are discussed, along with common MRI sequences and their utility.
Collapse
Affiliation(s)
- Catherine Joseph
- From Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
36
|
Di Salle G, Tumminello L, Laino ME, Shalaby S, Aghakhanyan G, Fanni SC, Febi M, Shortrede JE, Miccoli M, Faggioni L, Cosottini M, Neri E. Accuracy of Radiomics in Predicting IDH Mutation Status in Diffuse Gliomas: A Bivariate Meta-Analysis. Radiol Artif Intell 2024; 6:e220257. [PMID: 38231039 PMCID: PMC10831518 DOI: 10.1148/ryai.220257] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 09/12/2023] [Accepted: 10/24/2023] [Indexed: 01/18/2024]
Abstract
Purpose To perform a systematic review and meta-analysis assessing the predictive accuracy of radiomics in the noninvasive determination of isocitrate dehydrogenase (IDH) status in grade 4 and lower-grade diffuse gliomas. Materials and Methods A systematic search was performed in the PubMed, Scopus, Embase, Web of Science, and Cochrane Library databases for relevant articles published between January 1, 2010, and July 7, 2021. Pooled sensitivity and specificity across studies were estimated. Risk of bias was evaluated using Quality Assessment of Diagnostic Accuracy Studies-2, and methods were evaluated using the radiomics quality score (RQS). Additional subgroup analyses were performed according to tumor grade, RQS, and number of sequences used (PROSPERO ID: CRD42021268958). Results Twenty-six studies that included 3280 patients were included for analysis. The pooled sensitivity and specificity of radiomics for the detection of IDH mutation were 79% (95% CI: 76, 83) and 80% (95% CI: 76, 83), respectively. Low RQS scores were found overall for the included works. Subgroup analyses showed lower false-positive rates in very low RQS studies (RQS < 6) (meta-regression, z = -1.9; P = .02) compared with adequate RQS studies. No substantial differences were found in pooled sensitivity and specificity for the pure grade 4 gliomas group compared with the all-grade gliomas group (81% and 86% vs 79% and 79%, respectively) and for studies using single versus multiple sequences (80% and 77% vs 79% and 82%, respectively). Conclusion The pooled data showed that radiomics achieved good accuracy performance in distinguishing IDH mutation status in patients with grade 4 and lower-grade diffuse gliomas. The overall methodologic quality (RQS) was low and introduced potential bias. Keywords: Neuro-Oncology, Radiomics, Integration, Application Domain, Glioblastoma, IDH Mutation, Radiomics Quality Scoring Supplemental material is available for this article. Published under a CC BY 4.0 license.
Collapse
Affiliation(s)
- Gianfranco Di Salle
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Lorenzo Tumminello
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Maria Elena Laino
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Sherif Shalaby
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Gayane Aghakhanyan
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Salvatore Claudio Fanni
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Maria Febi
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Jorge Eduardo Shortrede
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Mario Miccoli
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Lorenzo Faggioni
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Mirco Cosottini
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| | - Emanuele Neri
- From Academic Radiology, Department of Translational Research on New
Technologies in Medicine and Surgery (G.D.S., L.T., G.A., S.C.F., M.F., J.E.S.,
L.F., E.N.), Department of Clinical and Experimental Medicine (M.M.), and
Neuroradiology Unit, Department of Translational Research on New Technologies in
Medicine and Surgery (M.C.), University of Pisa, Via Roma 67, 56126 Pisa, Italy;
Artificial Intelligence Center, IRCCS Humanitas Research Hospital, Rozzano,
Milan, Italy (M.E.L.); The Shrewsbury and Telford Hospital NHS Trust,
Shrewsbury, England (S.S.); and Italian Society of Medical and Interventional
Radiology, SIRM Foundation, Milan, Italy (E.N.)
| |
Collapse
|
37
|
Ullah A, Ullah S, Waqas M, Khan M, Rehman NU, Khalid A, Jan A, Aziz S, Naeem M, Halim SA, Khan A, Al-Harrasi A. Novel Natural Inhibitors for Glioblastoma by Targeting Epidermal Growth Factor Receptor and Phosphoinositide 3-kinase. Curr Med Chem 2024; 31:6596-6613. [PMID: 38616761 DOI: 10.2174/0109298673293279240404080046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND/AIM Glioblastoma is an extensively malignant neoplasm of the brain that predominantly impacts the human population. To address the challenge of glioblastoma, herein, we have searched for new drug-like candidates by extensive computational and biochemical investigations. METHODS Approximately 950 compounds were virtually screened against the two most promising targets of glioblastoma, i.e., epidermal growth factor receptor (EGFR) and phosphoinositide 3-kinase (PI3K). Based on highly negative docking scores, excellent binding capabilities and good pharmacokinetic properties, eight and seven compounds were selected for EGFR and PI3K, respectively. RESULTS Among those hits, four natural products (SBEH-40, QUER, QTME-12, and HCFR) exerted dual inhibitory effects on EGFR and PI3K in our in-silico analysis; therefore, their capacity to suppress the cell proliferation was assessed in U87 cell line (type of glioma cell line). The compounds SBEH-40, QUER, and QTME-12 exhibited significant anti-proliferative capability with IC50 values of 11.97 ± 0.73 μM, 28.27 ± 1.52 μM, and 22.93 ± 1.63 μM respectively, while HCFR displayed weak inhibitory potency (IC50 = 74.97 ± 2.30 μM). CONCLUSION This study has identified novel natural products that inhibit the progression of glioblastoma; however, further examinations of these molecules are required in animal and tissue models to better understand their downstream targeting mechanisms.
Collapse
Affiliation(s)
- Atta Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Muhammad Waqas
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
- Department of Biotechnology and Genetic Engineering, Hazara University, Mansehra, Pakistan
| | - Majid Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Najeeb Ur Rehman
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan 45142, Saudi Arabia
| | - Afnan Jan
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Shahkaar Aziz
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar 25130, Pakistan
| | - Muhammad Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| |
Collapse
|
38
|
Klunko NS, Achmad H, Abdullah TM, Mohammed S, Saha I, Salim KS, Obaid RF, Romero-Parra RM, Al-Hasnawi SS, Al-Janabi WH, Farhood B. The Anti-hypoxia Potentials of Trans-sodium Crocetinate in Hypoxiarelated Diseases: A Review. Curr Radiopharm 2024; 17:30-37. [PMID: 37877507 DOI: 10.2174/0118744710268127231020083505] [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: 06/19/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023]
Abstract
Crocetin is a kind of apocarotenoid carboxylic acid extracted from saffron (Crocus sativus L.), which is effective in upregulating tissue oxygenation. However, crocetin is difficult to solubilize. It was shown that the trans isomer of crocetin is effective in improving oxygen diffusivity, while its cis isomer appears not to be. Hence, the isolated trans isomer of crocetin or trans-sodium crocetinate (TSC) can be used instead of crocetin. It is shown that TSC can upregulate hypoxic tissue oxygenation and be effective in treating some hypoxia-related diseases. Moreover, experimental and clinical studies have reported no adverse effects following TSC treatment, even at high doses. The current study will discuss the potential role of TSC in hemorrhagic shock, ischemia, brain tumor radiotherapy, and others.
Collapse
Affiliation(s)
- Natalia S Klunko
- Department of Training of Scientific and Scientific-Pedagogical Personnel, Russian New University, Moscow, Russia
| | - Harun Achmad
- Department of Pediatric Dentistry, Faculty of Dentistry, Hasanuddin University, Makassar, Indonesia
| | | | - Sami Mohammed
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Indranil Saha
- Department of Physics, GLA University, Mathura, Pin- 281406, U.P., India
| | | | - Rasha Fadhel Obaid
- Department of Biomedical Engineering, Al-Mustaqbal University College, Babylon, Iraq
| | | | | | | | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
39
|
Pflug KM, Lee DW, Tripathi A, Bankaitis VA, Burgess K, Sitcheran R. Cyanine Dye Conjugation Enhances Crizotinib Localization to Intracranial Tumors, Attenuating NF-κB-Inducing Kinase Activity and Glioma Progression. Mol Pharm 2023; 20:6140-6150. [PMID: 37939020 DOI: 10.1021/acs.molpharmaceut.3c00496] [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: 11/10/2023]
Abstract
Glioblastoma (GBM) is a highly aggressive form of brain cancer with a poor prognosis and limited treatment options. The ALK and c-MET inhibitor Crizotinib has demonstrated preclinical therapeutic potential for newly diagnosed GBM, although its efficacy is limited by poor penetration of the blood brain barrier. Here, we identify Crizotinib as a novel inhibitor of nuclear factor-κB (NF-κB)-inducing kinase, which is a key regulator of GBM growth and proliferation. We further show that the conjugation of Crizotinib to a heptamethine cyanine dye, or a near-infrared dye (IR-Crizotinib), attenuated glioma cell proliferation and survival in vitro to a greater extent than unconjugated Crizotinib. Moreover, we observed increased IR-Crizotinib localization to orthotopic mouse xenograft GBM tumors, which resulted in impaired tumor growth in vivo. Overall, IR-Crizotinib exhibited improved intracranial chemotherapeutic delivery and tumor localization with concurrent inhibition of NIK and noncanonical NF-κB signaling, thereby reducing glioma growth in vitro, as well as in vivo, and increasing survival in a preclinical rodent model.
Collapse
Affiliation(s)
- Kathryn M Pflug
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Dong W Lee
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Ashutosh Tripathi
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Vytas A Bankaitis
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| | - Kevin Burgess
- Department of Chemistry, Texas A&M University, Box 30012, College Station, Texas 77842, United States
| | - Raquel Sitcheran
- Department of Cellular Biology and Genetics, Texas A&M University Health Science Center , College Station, Texas 77807, United States
| |
Collapse
|
40
|
Ramos-Fresnedo A, Al-Kharboosh R, Twohy EL, Basil AN, Szymkiewicz EC, Zubair AC, Trifiletti DM, Durand N, Dickson DW, Middlebrooks EH, Abarbanel DN, Tzeng SY, Almeida JP, Chaichana KL, Green JJ, Sherman WJ, Quiñones-Hinojosa A. Phase 1, Dose Escalation, Nonrandomized, Open-Label, Clinical Trial Evaluating the Safety and Preliminary Efficacy of Allogenic Adipose-Derived Mesenchymal Stem Cells for Recurrent Glioblastoma: A Clinical Trial Protocol. NEUROSURGERY PRACTICE 2023; 4:e00062. [PMID: 38464470 PMCID: PMC10923529 DOI: 10.1227/neuprac.0000000000000062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 02/18/2025]
Abstract
Background and Objectives Despite standard of care with maximal safe resection and chemoradiation, glioblastoma is the most common and aggressive type of primary brain cancer. Surgical resection provides a window of opportunity to locally treat gliomas while the patient is recovering, and before initiating concomitant chemoradiation. To assess the safety and establish the maximum tolerated dose of adipose-derived mesenchymal stem cells (AMSCs) for the treatment of recurrent glioblastoma (GBM). Secondary objectives are to assess the toxicity profile and long-term survival outcomes of patients enrolled in the trial. Additionally, biospecimens will be collected to explore the local and systemic responses to this therapy. Methods We will conduct a phase 1, dose escalated, non-randomized, open label, clinical trial of GBM patients who are undergoing surgical resection for recurrence. Up to 18 patients will receive intra-cavitary application of AMSCs encapsulated in fibrin glue during surgical resection. All patients will be followed for up to 5 years for safety and survival data. Adverse events will be recorded using the CTCAE V5.0. Expected Outcomes This study will explore the maximum tolerated dose (MTD) of AMSCs along with the toxicity profile of this therapy in patients with recurrent GBM. Additionally, preliminary long-term survival and progression-free survival outcome analysis will be used to power further randomized studies. Lastly, CSF and blood will be obtained throughout the treatment period to investigate circulating molecular and inflammatory tumoral/stem cell markers and explore the mechanism of action of the therapeutic intervention. Discussion This prospective translational study will determine the initial safety and toxicity profile of local delivery of AMSCs for recurrent GBM. It will also provide additional survival metrics for future randomized trials.
Collapse
Affiliation(s)
| | | | - Erin L. Twohy
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Abba C. Zubair
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Nisha Durand
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida, USA
| | - Dennis W. Dickson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
| | - Erik H. Middlebrooks
- Department of Radiology, Neuroradiology Division, Mayo Clinic, Jacksonville, Florida, USA
| | - David N. Abarbanel
- Department of Neurology, Neuro-Oncology Division, Mayo Clinic, Jacksonville, Florida, USA
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | - Jordan J. Green
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Wendy J. Sherman
- Department of Neurology, Neuro-Oncology Division, Mayo Clinic, Jacksonville, Florida, USA
| | | |
Collapse
|
41
|
José-López R. Chemotherapy for the treatment of intracranial glioma in dogs. Front Vet Sci 2023; 10:1273122. [PMID: 38026627 PMCID: PMC10643662 DOI: 10.3389/fvets.2023.1273122] [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: 08/05/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
Gliomas are the second most common primary brain tumor in dogs and although they are associated with a poor prognosis, limited data are available relating to the efficacy of standard therapeutic options such as surgery, radiation and chemotherapy. Additionally, canine glioma is gaining relevance as a naturally occurring animal model that recapitulates human disease with fidelity. There is an intense comparative research drive to test new therapeutic approaches in dogs and assess if results translate efficiently into human clinical trials to improve the poor outcomes associated with the current standard-of-care. However, the paucity of data and controversy around most appropriate treatment for intracranial gliomas in dogs make comparisons among modalities troublesome. To further inform therapeutic decision-making, client discussion, and future studies evaluating treatment responses, the outcomes of 127 dogs with intracranial glioma, either presumed (n = 49) or histologically confirmed (n = 78), that received chemotherapy as leading or adjuvant treatment are reviewed here. This review highlights the status of current chemotherapeutic approaches to intracranial gliomas in dogs, most notably temozolomide and lomustine; areas of novel treatment currently in development, and difficulties to consensuate and compare different study observations. Finally, suggestions are made to facilitate evidence-based research in the field of canine glioma therapeutics.
Collapse
Affiliation(s)
- Roberto José-López
- Hamilton Specialist Referrals – IVC Evidensia, High Wycombe, United Kingdom
| |
Collapse
|
42
|
Strack C, Pomykala KL, Schlemmer HP, Egger J, Kleesiek J. "A net for everyone": fully personalized and unsupervised neural networks trained with longitudinal data from a single patient. BMC Med Imaging 2023; 23:174. [PMID: 37907876 PMCID: PMC10619304 DOI: 10.1186/s12880-023-01128-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 10/16/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND With the rise in importance of personalized medicine and deep learning, we combine the two to create personalized neural networks. The aim of the study is to show a proof of concept that data from just one patient can be used to train deep neural networks to detect tumor progression in longitudinal datasets. METHODS Two datasets with 64 scans from 32 patients with glioblastoma multiforme (GBM) were evaluated in this study. The contrast-enhanced T1w sequences of brain magnetic resonance imaging (MRI) images were used. We trained a neural network for each patient using just two scans from different timepoints to map the difference between the images. The change in tumor volume can be calculated with this map. The neural networks were a form of a Wasserstein-GAN (generative adversarial network), an unsupervised learning architecture. The combination of data augmentation and the network architecture allowed us to skip the co-registration of the images. Furthermore, no additional training data, pre-training of the networks or any (manual) annotations are necessary. RESULTS The model achieved an AUC-score of 0.87 for tumor change. We also introduced a modified RANO criteria, for which an accuracy of 66% can be achieved. CONCLUSIONS We show a novel approach to deep learning in using data from just one patient to train deep neural networks to monitor tumor change. Using two different datasets to evaluate the results shows the potential to generalize the method.
Collapse
Affiliation(s)
- Christian Strack
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Girardetstraße 2, 45131, Essen, Germany.
- Division of Radiology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
- Medical Faculty Heidelberg, Heidelberg University, 69120, Heidelberg, Germany.
| | - Kelsey L Pomykala
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Girardetstraße 2, 45131, Essen, Germany
| | - Heinz-Peter Schlemmer
- Division of Radiology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Jan Egger
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Girardetstraße 2, 45131, Essen, Germany
- Cancer Research Center Cologne Essen (CCCE), University Medicine Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Jens Kleesiek
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Girardetstraße 2, 45131, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen, Hufelandstraße 55, 45147, Essen, Germany
- Cancer Research Center Cologne Essen (CCCE), University Medicine Essen, Hufelandstraße 55, 45147, Essen, Germany
- Department of Physics, TU Dortmund University, Otto-Hahn-Straße 4, D-44227, Dortmund, Germany
| |
Collapse
|
43
|
Mohammad AH, Jatana S, Ruiz-Barerra MA, Khalaf R, Al-Saadi T, Diaz RJ. Metformin use is associated with longer survival in glioblastoma patients with MGMT gene silencing. J Neurooncol 2023; 165:209-218. [PMID: 37889443 DOI: 10.1007/s11060-023-04485-2] [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: 09/25/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023]
Abstract
PURPOSE New treatments are needed to improve the overall survival of patients with glioblastoma Metformin is known for anti-tumorigenic effects in cancers, including breast and pancreas cancers. In this study, we assessed the association between metformin use and overall survival in glioblastoma patients. METHODS We retrospectively studied 241 patients who underwent surgery at diagnosis of glioblastoma between 2014 and 2018. Metformin was used for pre-existing type 2 diabetes mellitus or in the prevention or management of glucocorticoid induced hyperglycemia. Kaplan-Meier curves and log-rank p test were used for univariate analysis. Cox-proportional hazards model was used to generate adjusted hazard ratios for multivariate analysis. RESULTS Metformin use was associated with longer survival in patients with tumors that had a methylated O6-methylguanine DNA methyltransferase gene (MGMT) promoter (484 days 95% CI: 56-911 vs. 394 days 95% CI: 249-538, Log-Rank test: 6.5, p = 0.01). Cox regression analysis shows that metformin associates with lower risk of death at 2 years in patients with glioblastoma containing a methylated MGMT promoter (aHR = 0.497, 95% CI 0.26-0.93, p = 0.028). CONCLUSION Our findings suggest a survival benefit with metformin use in patients with glioblastomas having methylation of the MGMT promoter.
Collapse
Affiliation(s)
| | | | - Miguel Angel Ruiz-Barerra
- Neuro-Oncology Research Group, National Institute of Cancer, Bogotá, Colombia
- Department of Neurosurgery, National Institute of Cancer, Bogotá, Colombia
| | - Roy Khalaf
- Faculty of Medicine, McGill University, Montreal, Canada
| | - Tariq Al-Saadi
- Faculty of Medicine, McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute - McGill University Health Centre, Montreal, Canada
| | - Roberto J Diaz
- Faculty of Medicine, McGill University, Montreal, Canada.
- Department of Neurology and Neurosurgery, Montreal Neurological Institute - McGill University Health Centre, Montreal, Canada.
- Neurosurgical Oncology, Department of Neurology and Neurosurgery, Montreal Neurological Hospital - McGill University Health Centre, Montreal, Canada.
- Neurosurgical Oncology, Department of Neurology and Neurosurgery, Montreal Neurological Hospital, Faculty of Medicine, Faculty of Medicine, McGill University, 3801 University Street, Montreal, QC, H3A 2B4, Canada.
| |
Collapse
|
44
|
Bartusik-Aebisher D, Serafin I, Dynarowicz K, Aebisher D. Photodynamic therapy and associated targeting methods for treatment of brain cancer. Front Pharmacol 2023; 14:1250699. [PMID: 37841921 PMCID: PMC10568033 DOI: 10.3389/fphar.2023.1250699] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Brain tumors, including glioblastoma multiforme, are currently a cause of suffering and death of tens of thousands of people worldwide. Despite advances in clinical treatment, the average patient survival time from the moment of diagnosis of glioblastoma multiforme and application of standard treatment methods such as surgical resection, radio- and chemotherapy, is less than 4 years. The continuing development of new therapeutic methods for targeting and treating brain tumors may extend life and provide greater comfort to patients. One such developing therapeutic method is photodynamic therapy. Photodynamic therapy is a progressive method of therapy used in dermatology, dentistry, ophthalmology, and has found use as an antimicrobial agent. It has also found wide application in photodiagnosis. Photodynamic therapy requires the presence of three necessary components: a clinically approved photosensitizer, oxygen and light. This paper is a review of selected literature from Pubmed and Scopus scientific databases in the field of photodynamic therapy in brain tumors with an emphasis on glioblastoma treatment.
Collapse
Affiliation(s)
- Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - Iga Serafin
- Students English Division Science Club, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, Rzeszów, Poland
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of the University of Rzeszów, Rzeszów, Poland
| |
Collapse
|
45
|
Cook AB, Palange A, Schlich M, Bellotti E, Brahmachari S, di Francesco M, Decuzzi P. Matrix metalloproteinase responsive hydrogel microplates for programmed killing of invasive tumour cells. RSC APPLIED POLYMERS 2023; 1:19-29. [PMID: 38013908 PMCID: PMC10540463 DOI: 10.1039/d3lp00057e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/08/2023] [Indexed: 11/29/2023]
Abstract
Interactive materials are an emerging class of systems that can offer control over response and adaptivity in polymer structures towards the meso- and macroscale. Here, we use enzyme regulated cleavage of peptide crosslinkers in polymer hydrogels to release a cytotoxic therapeutic nanoparticle with an adaptable mechanism. Hydrogel microplates were formed through polyethylene glycol/peptide photoinitiated thiol-ene chemistry in a soft-lithography process to give square plates of 20 by 20 μm with a height of 10 μm. The peptide was chosen to be degradable in the presence of matrix metalloproteinase 2/9 (MMP-2/9). The hydrogel material's mechanical properties, swelling, and protease degradation were characterised. The microfabricated hydrogels were loaded with docetaxel (DTXL) containing poly(dl-lactide-co-glycolide) (PLGA) nanoparticles, and characterised for enzyme responsivity, and toxicity to MMP-2/9 overexpressing brain cancer cell line U87-MG. A 5-fold decrease in EC50 was seen compared to free DTXL, and a 20-fold decrease was seen for the MMP responsive microplates versus a non-degradable control microplate. Potential applications of this system in post-resection glioblastoma treatment are envisioned.
Collapse
Affiliation(s)
- Alexander B Cook
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia Via Morego 16163 Genova Italy
| | - Annalisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia Via Morego 16163 Genova Italy
| | - Michele Schlich
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia Via Morego 16163 Genova Italy
| | - Elena Bellotti
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia Via Morego 16163 Genova Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia Via Morego 16163 Genova Italy
| | - Martina di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia Via Morego 16163 Genova Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia Via Morego 16163 Genova Italy
| |
Collapse
|
46
|
Repici A, Ardizzone A, Filippone A, Colarossi C, Mare M, Raciti G, Mannino D, Cuzzocrea S, Paterniti I, Esposito E. Interleukin-21 Influences Glioblastoma Course: Biological Mechanisms and Therapeutic Potential. Cells 2023; 12:2284. [PMID: 37759505 PMCID: PMC10526836 DOI: 10.3390/cells12182284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Brain tumors represent a heterogeneous group of neoplasms involving the brain or nearby tissues, affecting populations of all ages with a high incidence worldwide. Among the primary brain tumors, the most aggressive and also the most common is glioblastoma (GB), a type of glioma that falls into the category of IV-grade astrocytoma. GB often leads to death within a few months after diagnosis, even if the patient is treated with available therapies; for this reason, it is important to continue to discover new therapeutic approaches to allow for a better survival rate of these patients. Immunotherapy, today, seems to be one of the most innovative types of treatment, based on the ability of the immune system to counteract various pathologies, including cancer. In this context, interleukin 21 (IL-21), a type I cytokine produced by natural killer (NK) cells and CD4+ T lymphocytes, appears to be a valid target for new therapies since this cytokine is involved in the activation of innate and adaptive immunity. To match this purpose, our review deeply evaluated how IL-21 could influence the progression of GB, analyzing its main biological processes and mechanisms while evaluating the potential use of the latest available therapies.
Collapse
Affiliation(s)
- Alberto Repici
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Cristina Colarossi
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (C.C.); (M.M.)
| | - Marzia Mare
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (C.C.); (M.M.)
| | - Gabriele Raciti
- IOM Ricerca, Via Penninazzo 11, 95029 Viagrande, Italy;
- Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 98166 Messina, Italy; (A.R.); (A.A.); (A.F.); (D.M.); (S.C.); (E.E.)
| |
Collapse
|
47
|
Kawak P, Sawaftah NMA, Pitt WG, Husseini GA. Transferrin-Targeted Liposomes in Glioblastoma Therapy: A Review. Int J Mol Sci 2023; 24:13262. [PMID: 37686065 PMCID: PMC10488197 DOI: 10.3390/ijms241713262] [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: 06/19/2023] [Revised: 07/14/2023] [Accepted: 07/31/2023] [Indexed: 09/10/2023] Open
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor, and its treatment is further complicated by the high selectivity of the blood-brain barrier (BBB). The scientific community is urgently seeking innovative and effective therapeutic solutions. Liposomes are a promising new tool that has shown potential in addressing the limitations of chemotherapy, such as poor bioavailability and toxicity to healthy cells. However, passive targeting strategies based solely on the physicochemical properties of liposomes have proven ineffective due to a lack of tissue specificity. Accordingly, the upregulation of transferrin receptors (TfRs) in brain tissue has led to the development of TfR-targeted anticancer therapeutics. Currently, one of the most widely adopted methods for improving drug delivery in the treatment of GBM and other neurological disorders is the utilization of active targeting strategies that specifically target this receptor. In this review, we discuss the role of Tf-conjugated liposomes in GBM therapy and present some recent studies investigating the drug delivery efficiency of Tf-liposomes; in addition, we address some challenges currently facing this approach to treatment and present some potential improvement possibilities.
Collapse
Affiliation(s)
- Paul Kawak
- Chemical and Biological Engineering Department, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
| | - Nour M. Al Sawaftah
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
| | - William G. Pitt
- Chemical Engineering Department, Brigham Young University, Provo, UT 84602, USA
| | - Ghaleb A. Husseini
- Chemical and Biological Engineering Department, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates;
| |
Collapse
|
48
|
García-Montaño LA, Licón-Muñoz Y, Martinez FJ, Keddari YR, Ziemke MK, Chohan MO, Piccirillo SG. Dissecting Intra-tumor Heterogeneity in the Glioblastoma Microenvironment Using Fluorescence-Guided Multiple Sampling. Mol Cancer Res 2023; 21:755-767. [PMID: 37255362 PMCID: PMC10390891 DOI: 10.1158/1541-7786.mcr-23-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/25/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
The treatment of the most aggressive primary brain tumor in adults, glioblastoma (GBM), is challenging due to its heterogeneous nature, invasive potential, and poor response to chemo- and radiotherapy. As a result, GBM inevitably recurs and only a few patients survive 5 years post-diagnosis. GBM is characterized by extensive phenotypic and genetic heterogeneity, creating a diversified genetic landscape and a network of biological interactions between subclones, ultimately promoting tumor growth and therapeutic resistance. This includes spatial and temporal changes in the tumor microenvironment, which influence cellular and molecular programs in GBM and therapeutic responses. However, dissecting phenotypic and genetic heterogeneity at spatial and temporal levels is extremely challenging, and the dynamics of the GBM microenvironment cannot be captured by analysis of a single tumor sample. In this review, we discuss the current research on GBM heterogeneity, in particular, the utility and potential applications of fluorescence-guided multiple sampling to dissect phenotypic and genetic intra-tumor heterogeneity in the GBM microenvironment, identify tumor and non-tumor cell interactions and novel therapeutic targets in areas that are key for tumor growth and recurrence, and improve the molecular classification of GBM.
Collapse
Affiliation(s)
- Leopoldo A. García-Montaño
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yamhilette Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Frank J. Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yasine R. Keddari
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of California, Merced, California
| | - Michael K. Ziemke
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Muhammad O. Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sara G.M. Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| |
Collapse
|
49
|
D'Alessandris QG, Menna G, Izzo A, D'Ercole M, Della Pepa GM, Lauretti L, Pallini R, Olivi A, Montano N. Neuromodulation for Brain Tumors: Myth or Reality? A Narrative Review. Int J Mol Sci 2023; 24:11738. [PMID: 37511496 PMCID: PMC10380317 DOI: 10.3390/ijms241411738] [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: 05/31/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
In recent years, research on brain cancers has turned towards the study of the interplay between the tumor and its host, the normal brain. Starting from the establishment of a parallelism between neurogenesis and gliomagenesis, the influence of neuronal activity on the development of brain tumors, particularly gliomas, has been partially unveiled. Notably, direct electrochemical synapses between neurons and glioma cells have been identified, paving the way for new approaches for the cure of brain cancers. Since this novel field of study has been defined "cancer neuroscience", anticancer therapeutic approaches exploiting these discoveries can be referred to as "cancer neuromodulation". In the present review, we provide an up-to-date description of the novel findings and of the therapeutic neuromodulation perspectives in cancer neuroscience. We focus both on more traditional oncologic approaches, aimed at modulating the major pathways involved in cancer neuroscience through drugs or genetic engineering techniques, and on electric stimulation proposals; the latter is at the cutting-edge of neuro-oncology.
Collapse
Affiliation(s)
- Quintino Giorgio D'Alessandris
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Grazia Menna
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
| | - Alessandro Izzo
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Manuela D'Ercole
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Giuseppe Maria Della Pepa
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Liverana Lauretti
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Roberto Pallini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Alessandro Olivi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Nicola Montano
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
50
|
Das A, Ding S, Liu R, Huang C. Quantifying the Growth of Glioblastoma Tumors Using Multimodal MRI Brain Images. Cancers (Basel) 2023; 15:3614. [PMID: 37509277 PMCID: PMC10377296 DOI: 10.3390/cancers15143614] [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: 06/19/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Predicting the eventual volume of tumor cells, that might proliferate from a given tumor, can help in cancer early detection and medical procedure planning to prevent their migration to other organs. In this work, a new statistical framework is proposed using Bayesian techniques for detecting the eventual volume of cells expected to proliferate from a glioblastoma (GBM) tumor. Specifically, the tumor region was first extracted using a parallel image segmentation algorithm. Once the tumor region was determined, we were interested in the number of cells that could proliferate from this tumor until its survival time. For this, we constructed the posterior distribution of the tumor cell numbers based on the proposed likelihood function and a certain prior volume. Furthermore, we extended the detection model and conducted a Bayesian regression analysis by incorporating radiomic features to discover those non-tumor cells that remained undetected. The main focus of the study was to develop a time-independent prediction model that could reliably predict the ultimate volume a malignant tumor of the fourth-grade severity could attain and which could also determine if the incorporation of the radiomic properties of the tumor enhanced the chances of no malignant cells remaining undetected.
Collapse
Affiliation(s)
- Anisha Das
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Shengxian Ding
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Rongjie Liu
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Chao Huang
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|