51
|
Ou M, Cho HY, Fu J, Thein TZ, Wang W, Swenson SD, Minea RO, Stathopoulos A, Schönthal AH, Hofman FM, Tang L, Chen TC. Inhibition of autophagy and induction of glioblastoma cell death by NEO214, a perillyl alcohol-rolipram conjugate. Autophagy 2023; 19:3169-3188. [PMID: 37545052 PMCID: PMC10621246 DOI: 10.1080/15548627.2023.2242696] [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: 02/06/2022] [Accepted: 07/26/2023] [Indexed: 08/08/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor, exhibiting a high rate of recurrence and poor prognosis. Surgery and chemoradiation with temozolomide (TMZ) represent the standard of care, but, in most cases, the tumor develops resistance to further treatment and the patients succumb to disease. Therefore, there is a great need for the development of well-tolerated, effective drugs that specifically target chemoresistant gliomas. NEO214 was generated by covalently conjugating rolipram, a PDE4 (phosphodiesterase 4) inhibitor, to perillyl alcohol, a naturally occurring monoterpene related to limonene. Our previous studies in preclinical models showed that NEO214 harbors anticancer activity, is able to cross the blood-brain barrier (BBB), and is remarkably well tolerated. In the present study, we investigated its mechanism of action and discovered inhibition of macroautophagy/autophagy as a key component of its anticancer effect in glioblastoma cells. We show that NEO214 prevents autophagy-lysosome fusion, thereby blocking autophagic flux and triggering glioma cell death. This process involves activation of MTOR (mechanistic target of rapamycin kinase) activity, which leads to cytoplasmic accumulation of TFEB (transcription factor EB), a critical regulator of genes involved in the autophagy-lysosomal pathway, and consequently reduced expression of autophagy-lysosome genes. When combined with chloroquine and TMZ, the anticancer impact of NEO214 is further potentiated and unfolds against TMZ-resistant cells as well. Taken together, our findings characterize NEO214 as a novel autophagy inhibitor that could become useful for overcoming chemoresistance in glioblastoma.Abbreviations: ATG: autophagy related; BAFA1: bafilomycin A1; BBB: blood brain barrier; CQ: chloroquine; GBM: glioblastoma; LAMP1: lysosomal associated membrane protein 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MGMT: O-6-methylguanine-DNA methyltransferase; MTOR: mechanistic target of rapamycin kinase; MTORC: MTOR complex; POH: perillyl alcohol; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB; TMZ: temozolomide.
Collapse
Affiliation(s)
- Mengting Ou
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Hee-Yeon Cho
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Chemistry, Physics, and Engineering, Biola University, La Mirada, CA, USA
| | - Jie Fu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Thu Zan Thein
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Weijun Wang
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stephen D. Swenson
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Radu O. Minea
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Apostolos Stathopoulos
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Axel H. Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Florence M. Hofman
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Thomas C. Chen
- Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
52
|
Chinyama HA, Wei L, Mokgautsi N, Lawal B, Wu ATH, Huang HS. Identification of CDK1, PBK, and CHEK1 as an Oncogenic Signature in Glioblastoma: A Bioinformatics Approach to Repurpose Dapagliflozin as a Therapeutic Agent. Int J Mol Sci 2023; 24:16396. [PMID: 38003585 PMCID: PMC10671581 DOI: 10.3390/ijms242216396] [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: 09/25/2023] [Revised: 10/27/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and lethal primary brain tumor whose median survival is less than 15 months. The current treatment regimen comprising surgical resectioning, chemotherapy with Temozolomide (TMZ), and adjuvant radiotherapy does not achieve total patient cure. Stem cells' presence and GBM tumor heterogeneity increase their resistance to TMZ, hence the poor overall survival of patients. A dysregulated cell cycle in glioblastoma enhances the rapid progression of GBM by evading senescence or apoptosis through an over-expression of cyclin-dependent kinases and other protein kinases that are the cell cycle's main regulatory proteins. Herein, we identified and validated the biomarker and predictive properties of a chemoradio-resistant oncogenic signature in GBM comprising CDK1, PBK, and CHEK1 through our comprehensive in silico analysis. We found that CDK1/PBK/CHEK1 overexpression drives the cell cycle, subsequently promoting GBM tumor progression. In addition, our Kaplan-Meier survival estimates validated the poor patient survival associated with an overexpression of these genes in GBM. We used in silico molecular docking to analyze and validate our objective to repurpose Dapagliflozin against CDK1/PBK/CHEK1. Our results showed that Dapagliflozin forms putative conventional hydrogen bonds with CDK1, PBK, and CHEK1 and arrests the cell cycle with the lowest energies as Abemaciclib.
Collapse
Affiliation(s)
- Harold A. Chinyama
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Li Wei
- Department of Neurosurgery, Wan Fang Hospital, Taipei Medical University, No.111, Sec. 3, Xinglong Rd., Taipei 11696, Taiwan;
- Taipei Neuroscience Institute, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei 11031, Taiwan
| | - Ntlotlang Mokgautsi
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan;
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Bashir Lawal
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - Alexander T. H. Wu
- PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan;
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- School of Pharmacy, National Defense Medical Center, Taipei 11490, Taiwan
- PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
53
|
Yu L, Lee KW, Zhao YQ, Xu Y, Zhou Y, Li M, Kim JS. Metal Modulation: An Effortless Tactic for Refining Photoredox Catalysis in Living Cells. Inorg Chem 2023; 62:18767-18778. [PMID: 37905835 DOI: 10.1021/acs.inorgchem.3c03284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The remarkable impact of photoredox catalytic chemistries has sparked a wave of innovation, opening doors to novel biotechnologies in the realm of catalytic antitumor therapy. Yet, the quest for novel photoredox catalysts (PCs) suitable for living systems, or the enhancement of catalytic efficacy in existing biocompatible PC systems, persists as a formidable challenge. Within this context, we introduce a readily applicable metal modulation strategy that significantly augments photoredox catalysis within living cells, exemplified by a set of metalloporphyrin complexes termed M-TCPPs (M = Zn, Mn, Ni, Co, Cu). Among these complexes, Zn-TCPP emerges as an exceptional catalyst, displaying remarkable photocatalytic activity in the oxidation of nicotinamide adenine dinucleotide (NADH), nicotinamide adenine dinucleotide phosphate (NADPH), and specific amino acids. Notably, comprehensive investigations reveal that Zn-TCPP's superior catalytic prowess primarily arises from the establishment of an efficient oxidative cycle for PC, in contrast to previously reported PCs engaged in reductive cycles. Moreover, theoretical calculations illuminate that amplified intersystem crossing rates and geometry alterations in Zn-TCPP contribute to its heightened photocatalytic performance. In vitro studies demonstrated that Zn-TCPP exhibits therapeutic potential and is found to be effective for photocatalytic antitumor therapy in both glioblastoma G98T cells and 3D multicellular spheroids. This study underscores the transformative role of "metal modulation" in advancing high-performance PCs for catalytic antitumor therapy, marking a significant stride toward the realization of this innovative therapeutic approach.
Collapse
Affiliation(s)
- Le Yu
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Kyung-Woo Lee
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Yu-Qiang Zhao
- College of Chemical Science and Technology, Yunnan University, Kunming 650091, China
| | - Yunjie Xu
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Ying Zhou
- College of Chemical Science and Technology, Yunnan University, Kunming 650091, China
| | - Mingle Li
- State Key Laboratory of Fine Chemicals, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
- TheranoChem Incorporation, Seongbuk-gu, Seoul 02856, Republic of Korea
| |
Collapse
|
54
|
Hu M, Liao X, Tao Y, Chen Y. Advances in oncolytic herpes simplex virus and adenovirus therapy for recurrent glioma. Front Immunol 2023; 14:1285113. [PMID: 38022620 PMCID: PMC10652401 DOI: 10.3389/fimmu.2023.1285113] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Recurrent glioma treatment is challenging due to molecular heterogeneity and treatment resistance commonly observed in these tumors. Researchers are actively pursuing new therapeutic strategies. Oncolytic viruses have emerged as a promising option. Oncolytic viruses selectively replicate within tumor cells, destroying them and stimulating the immune system for an enhanced anticancer response. Among Oncolytic viruses investigated for recurrent gliomas, oncolytic herpes simplex virus and oncolytic adenovirus show notable potential. Genetic modifications play a crucial role in optimizing their therapeutic efficacy. Different generations of replicative conditioned oncolytic human adenovirus and oncolytic HSV have been developed, incorporating specific modifications to enhance tumor selectivity, replication efficiency, and immune activation. This review article summarizes these genetic modifications, offering insights into the underlying mechanisms of Oncolytic viruses' therapy. It also aims to identify strategies for further enhancing the therapeutic benefits of Oncolytic viruses. However, it is important to acknowledge that additional research and clinical trials are necessary to establish the safety, efficacy, and optimal utilization of Oncolytic viruses in treating recurrent glioblastoma.
Collapse
Affiliation(s)
- Mingming Hu
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - XuLiang Liao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Tao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yaohui Chen
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
55
|
Ong K, Hounjet C, Makarenko S. Intraoperative and postoperative complications for repeat high-grade glioma resections with concurrent chemotherapy: patient series. JOURNAL OF NEUROSURGERY. CASE LESSONS 2023; 6:CASE23341. [PMID: 37870760 PMCID: PMC10584083 DOI: 10.3171/case23341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/25/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND High-grade gliomas are aggressive primary brain tumors, the most common of which is glioblastoma multiforme. Despite advances in treatment, the prognosis for these patients remains poor. The most common chemotherapeutic agents used in the treatment of this pathology include temozolomide (TMZ), procarbazine, lomustine, and vincristine. It is unclear whether chemotherapy should be held during resection for high-grade gliomas, because the perioperative risk profile is not clearly defined. OBSERVATIONS The authors report a case series of 18 surgeries to investigate the effects of concurrent TMZ and lomustine chemotherapy on surgical complications in patients undergoing repeat resection for recurrent high-grade gliomas. The authors found no postoperative infections, self-limiting postoperative complications, or excessive intraoperative blood loss and found one intraoperative complication. LESSONS There may not be a need to pause TMZ and lomustine chemotherapy during recurrent resections for high-grade gliomas, and continuing these medications throughout the perioperative period may be appropriate. This case series suggests that patients receiving TMZ and lomustine chemotherapy who need a repeat resection for recurrent high-grade gliomas should consider remaining on their chemotherapy regimen because it has been shown in the literature to improve recurrence-free survival time.
Collapse
Affiliation(s)
- Kenneth Ong
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Celine Hounjet
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; and
- Division of Neurosurgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Serge Makarenko
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; and
- Division of Neurosurgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
56
|
Tao Y, Zhu J, Yu X, Cong H, Li J, Cai T, Chen Q. Prognostic risk of immune-associated signature in the microenvironment of brain gliomas. Front Genet 2023; 14:1208651. [PMID: 37867596 PMCID: PMC10587408 DOI: 10.3389/fgene.2023.1208651] [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: 04/19/2023] [Accepted: 09/15/2023] [Indexed: 10/24/2023] Open
Abstract
Understanding the key factors in the tumor microenvironment (TME) that affect the prognosis of gliomas is crucial. In this study, we sought to uncover the prognostic significance of immune cells and immune-related genes in the TME of gliomas. We incorporated data of 970 glioma patient samples from the Chinese Glioma Genome Atlas (CGGA) database as the training set, and an additional set of 666 samples from The Cancer Genome Atlas (TCGA) database served as the validation set. From our analysis, we identified 21 immune-related differentially expressed genes (DEGs) in the TME, which holds implications for glioma prognosis. Based on these genes, we constructed a prognostic risk model on the 21 genes. The prognostic risk model demonstrated robust performance with an area under the curve (AUC) value of 0.848. Notably, the risk score derived from the model emerged as an independent prognostic factor of gliomas, with high risk scores indicative of an unfavorable prognosis. Furthermore, we observed that high infiltration levels of certain immune cells, namely, activated dendritic cells, M0 macrophages, M2 macrophages, and regulatory T cells (Tregs), correlated with an unfavorable glioma prognosis. In conclusion, our findings suggested that the TME of gliomas harbored a distinct immune-associated signature, comprising 21 immune-related genes and specific immune cells. These elements significantly influence the prognosis and present potential as novel indicators in the clinical assessment of glioma patient outcomes.
Collapse
Affiliation(s)
- Yaling Tao
- Ningbo No 2 Hospital, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | | | - Xiaoling Yu
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Huaiwei Cong
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Jinpeng Li
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Ting Cai
- Ningbo No 2 Hospital, Ningbo, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Qian Chen
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
- Thorgene Co., Ltd., Beijing, China
- Ningbo Hangzhou Bay Hospital, Ningbo, China
| |
Collapse
|
57
|
Faisal SM, Castro MG, Lowenstein PR. Combined cytotoxic and immune-stimulatory gene therapy using Ad-TK and Ad-Flt3L: Translational developments from rodents to glioma patients. Mol Ther 2023; 31:2839-2860. [PMID: 37574780 PMCID: PMC10556227 DOI: 10.1016/j.ymthe.2023.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/14/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023] Open
Abstract
Gliomas are the most prevalent and devastating primary malignant brain tumors in adults. Despite substantial advances in understanding glioma biology, there have been no regulatory drug approvals in the US since bevacizumab in 2009 and tumor treating fields in 2011. Recent phase III clinical trials have failed to meet their prespecified therapeutic primary endpoints, highlighting the need for novel therapies. The poor prognosis of glioma patients, resistance to chemo-radiotherapy, and the immunosuppressive tumor microenvironment underscore the need for the development of novel therapies. Gene therapy-based immunotherapeutic strategies that couple the ability of the host immune system to specifically kill glioma cells and develop immunological memory have shown remarkable progress. Two adenoviral vectors expressing Ad-HSV1-TK/GCV and Ad-Flt3L have shown promising preclinical data, leading to FDA approval of a non-randomized, phase I open-label, first in human trial to test safety, cytotoxicity, and immune-stimulatory efficiency in high-grade glioma patients (NCT01811992). This review provides a thorough overview of immune-stimulatory gene therapy highlighting recent advancements, potential drawbacks, future directions, and recommendations for future implementation of clinical trials.
Collapse
Affiliation(s)
- Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Rogel Cancer Centre, University of Michigan Medical School, Ann Arbor, MI 48108, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Rogel Cancer Centre, University of Michigan Medical School, Ann Arbor, MI 48108, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Rogel Cancer Centre, University of Michigan Medical School, Ann Arbor, MI 48108, USA; Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48108, USA.
| |
Collapse
|
58
|
Schaufler A, Sanin AY, Sandalcioglu IE, Hartmann K, Croner RS, Perrakis A, Wartmann T, Boese A, Kahlert UD, Fischer I. Concept of a fully-implantable system to monitor tumor recurrence. Sci Rep 2023; 13:16362. [PMID: 37773315 PMCID: PMC10541913 DOI: 10.1038/s41598-023-43226-3] [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: 04/12/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023] Open
Abstract
Current treatment for glioblastoma includes tumor resection followed by radiation, chemotherapy, and periodic post-operative examinations. Despite combination therapies, patients face a poor prognosis and eventual recurrence, which often occurs at the resection site. With standard MRI imaging surveillance, histologic changes may be overlooked or misinterpreted, leading to erroneous conclusions about the course of adjuvant therapy and subsequent interventions. To address these challenges, we propose an implantable system for accurate continuous recurrence monitoring that employs optical sensing of fluorescently labeled cancer cells and is implanted in the resection cavity during the final stage of tumor resection. We demonstrate the feasibility of the sensing principle using miniaturized system components, optical tissue phantoms, and porcine brain tissue in a series of experimental trials. Subsequently, the system electronics are extended to include circuitry for wireless energy transfer and power management and verified through electromagnetic field, circuit simulations and test of an evaluation board. Finally, a holistic conceptual system design is presented and visualized. This novel approach to monitor glioblastoma patients is intended to early detect recurrent cancerous tissue and enable personalization and optimization of therapy thus potentially improving overall prognosis.
Collapse
Affiliation(s)
- Anna Schaufler
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- Department of Neurosurgery, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- INKA Health Tech Innovation Lab., Medical Faculty, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Ahmed Y Sanin
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - I Erol Sandalcioglu
- Department of Neurosurgery, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Karl Hartmann
- Department of Neurosurgery, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Roland S Croner
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Aristotelis Perrakis
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Thomas Wartmann
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Axel Boese
- INKA Health Tech Innovation Lab., Medical Faculty, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Ulf D Kahlert
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular - and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University Magdeburg, 39120, Magdeburg, Germany
| | - Igor Fischer
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225, Düsseldorf, Germany.
| |
Collapse
|
59
|
Müller Bark J, Trevisan França de Lima L, Zhang X, Broszczak D, Leo PJ, Jeffree RL, Chua B, Day BW, Punyadeera C. Proteome profiling of salivary small extracellular vesicles in glioblastoma patients. Cancer 2023; 129:2836-2847. [PMID: 37254878 PMCID: PMC10952188 DOI: 10.1002/cncr.34888] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 04/15/2023] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs) play a critical role in intercellular communication under physiological and pathological conditions, including cancer. EVs cargo reflects their cell of origin, suggesting their utility as biomarkers. EVs are detected in several biofluids, and their ability to cross the blood-brain barrier has highlighted their potential as prognostic and diagnostic biomarkers in gliomas, including glioblastoma (GBM). Studies have demonstrated the potential clinical utility of plasma-derived EVs in glioma. However, little is known about the clinical utility of saliva-derived EVs in GBM. METHODS Small EVs were isolated from whole mouth saliva of GBM patients pre- and postoperatively. Isolation was performed using differential centrifugation and/or ultracentrifugation. EVs were characterized by concentration, size, morphology, and EVs cell-surface protein markers. Protein cargo in EVs was profiled using mass spectrometry. RESULTS There were no statistically significant differences in size and concentration of EVs derived from pre- and post GBM patients' saliva samples. A higher number of proteins were detected in preoperative samples compared to postoperative samples. The authors found four highly abundant proteins (aldolase A, 14-3-3 protein ε, enoyl CoA hydratase 1, and transmembrane protease serine 11B) in preoperative saliva samples from GBM patients with poor outcomes. Functional enrichment analysis of pre- and postoperative saliva samples showed significant enrichment of several pathways, including those related to the immune system, cell cycle and programmed cell death. CONCLUSIONS This study, for the first time, demonstrates the feasibility of isolating and characterizing small EVs from pre- and postoperative saliva samples from GBM patients. Preliminary findings encourage further large cohort validation studies on salivary small EVs to evaluate prognosis in GBM.
Collapse
Affiliation(s)
- Juliana Müller Bark
- Centre for Biomedical TechnologiesThe School of Biomedical SciencesFaculty of HealthQueensland University of TechnologyBrisbaneQueenslandAustralia
- Saliva and Liquid Biopsy Translational LaboratoryGriffith Institute for Drug Discovery Griffith UniversityBrisbaneQueenslandAustralia
- Translational Research InstituteBrisbaneQueenslandAustralia
| | - Lucas Trevisan França de Lima
- Centre for Biomedical TechnologiesThe School of Biomedical SciencesFaculty of HealthQueensland University of TechnologyBrisbaneQueenslandAustralia
- Saliva and Liquid Biopsy Translational LaboratoryGriffith Institute for Drug Discovery Griffith UniversityBrisbaneQueenslandAustralia
- Translational Research InstituteBrisbaneQueenslandAustralia
- Gallipoli Medical Research InstituteGreenslopes Private HospitalBrisbaneQueenslandAustralia
| | - Xi Zhang
- Centre for Biomedical TechnologiesThe School of Biomedical SciencesFaculty of HealthQueensland University of TechnologyBrisbaneQueenslandAustralia
- Saliva and Liquid Biopsy Translational LaboratoryGriffith Institute for Drug Discovery Griffith UniversityBrisbaneQueenslandAustralia
- Translational Research InstituteBrisbaneQueenslandAustralia
| | - Daniel Broszczak
- School of Biomedical SciencesFaculty of HealthQueensland University of TechnologyBrisbaneQueenslandAustralia
| | - Paul J. Leo
- Translational Research InstituteBrisbaneQueenslandAustralia
- Translational Genomics GroupQueensland University of TechnologyTranslational Research InstituteWoolloongabbaQueenslandAustralia
| | - Rosalind L. Jeffree
- Faculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
- Kenneth G. Jamieson Department of NeurosurgeryRoyal Brisbane and Women's HospitalHerstonQueenslandAustralia
| | - Benjamin Chua
- Faculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
- Cancer Care ServicesRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Bryan W. Day
- Cell and Molecular Biology DepartmentSid Faithfull Brain Cancer LaboratoryQIMR Berghofer MRIBrisbaneQueenslandAustralia
| | - Chamindie Punyadeera
- Centre for Biomedical TechnologiesThe School of Biomedical SciencesFaculty of HealthQueensland University of TechnologyBrisbaneQueenslandAustralia
- Saliva and Liquid Biopsy Translational LaboratoryGriffith Institute for Drug Discovery Griffith UniversityBrisbaneQueenslandAustralia
- Menzies Health Institute (MHIQ)Griffith UniversityGold CoastQueenslandAustralia
| |
Collapse
|
60
|
Karschnia P, Dono A, Young JS, Juenger ST, Teske N, Häni L, Sciortino T, Mau CY, Bruno F, Nunez L, Morshed RA, Haddad AF, Weller M, van den Bent M, Beck J, Hervey-Jumper S, Molinaro AM, Tandon N, Rudà R, Vogelbaum MA, Bello L, Schnell O, Grau SJ, Chang SM, Berger MS, Esquenazi Y, Tonn JC. Prognostic evaluation of re-resection for recurrent glioblastoma using the novel RANO classification for extent of resection: A report of the RANO resect group. Neuro Oncol 2023; 25:1672-1685. [PMID: 37253096 PMCID: PMC10479742 DOI: 10.1093/neuonc/noad074] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND The value of re-resection in recurrent glioblastoma remains controversial as a randomized trial that specifies intentional incomplete resection cannot be justified ethically. Here, we aimed to (1) explore the prognostic role of extent of re-resection using the previously proposed Response Assessment in Neuro-Oncology (RANO) classification (based upon residual contrast-enhancing (CE) and non-CE tumor), and to (2) define factors consolidating the surgical effects on outcome. METHODS The RANO resect group retrospectively compiled an 8-center cohort of patients with first recurrence from previously resected glioblastomas. The associations of re-resection and other clinical factors with outcome were analyzed. Propensity score-matched analyses were constructed to minimize confounding effects when comparing the different RANO classes. RESULTS We studied 681 patients with first recurrence of Isocitrate Dehydrogenase (IDH) wild-type glioblastomas, including 310 patients who underwent re-resection. Re-resection was associated with prolonged survival even when stratifying for molecular and clinical confounders on multivariate analysis; ≤1 cm3 residual CE tumor was associated with longer survival than non-surgical management. Accordingly, "maximal resection" (class 2) had superior survival compared to "submaximal resection" (class 3). Administration of (radio-)chemotherapy in the absence of postoperative deficits augmented the survival associations of smaller residual CE tumors. Conversely, "supramaximal resection" of non-CE tumor (class 1) was not associated with prolonged survival but was frequently accompanied by postoperative deficits. The prognostic role of residual CE tumor was confirmed in propensity score analyses. CONCLUSIONS The RANO resect classification serves to stratify patients with re-resection of glioblastoma. Complete resection according to RANO resect classes 1 and 2 is prognostic.
Collapse
Affiliation(s)
- Philipp Karschnia
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Germany
| | - Antonio Dono
- Department of Neurosurgery, McGovern Medical School at UT Health Houston, Houston, Texas, USA
| | - Jacob S Young
- Department of Neurosurgery and Division of Neuro-Oncology, University of San Francisco, San Francisco, California, USA
| | | | - Nico Teske
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany
| | - Levin Häni
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Tommaso Sciortino
- Division of Neuro-Oncology, Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Christine Y Mau
- Department of Neuro-Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Francesco Bruno
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Italy
| | - Luis Nunez
- Department of Diagnostic and Interventional Imaging, McGovern Medical School at UT Health Houston, Houston, Texas, USA
| | - Ramin A Morshed
- Department of Neurosurgery and Division of Neuro-Oncology, University of San Francisco, San Francisco, California, USA
| | - Alexander F Haddad
- Department of Neurosurgery and Division of Neuro-Oncology, University of San Francisco, San Francisco, California, USA
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Martin van den Bent
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Juergen Beck
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Shawn Hervey-Jumper
- Department of Neurosurgery and Division of Neuro-Oncology, University of San Francisco, San Francisco, California, USA
| | - Annette M Molinaro
- Department of Neurosurgery and Division of Neuro-Oncology, University of San Francisco, San Francisco, California, USA
| | - Nitin Tandon
- Department of Neurosurgery, McGovern Medical School at UT Health Houston, Houston, Texas, USA
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Italy
| | | | - Lorenzo Bello
- Division of Neuro-Oncology, Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Oliver Schnell
- Department of Neurosurgery, University of Freiburg, Freiburg, Germany
| | - Stefan J Grau
- Department of Neurosurgery, University of Cologne, Cologne, Germany
- Klinikum Fulda, Academic Hospital of Marburg University, Klinikum, Fulda, Germany
| | - Susan M Chang
- Department of Neurosurgery and Division of Neuro-Oncology, University of San Francisco, San Francisco, California, USA
| | - Mitchel S Berger
- Department of Neurosurgery and Division of Neuro-Oncology, University of San Francisco, San Francisco, California, USA
| | - Yoshua Esquenazi
- Department of Neurosurgery, McGovern Medical School at UT Health Houston, Houston, Texas, USA
| | - Joerg-Christian Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Germany
| |
Collapse
|
61
|
Zheng Y, Saffari SE, Low DCY, Lin X, Ker JRX, Ang SYL, Ng WH, Wan KR. Lobectomy versus gross total resection for glioblastoma multiforme: A systematic review and individual-participant data meta-analysis. J Clin Neurosci 2023; 115:60-65. [PMID: 37487449 DOI: 10.1016/j.jocn.2023.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/26/2023]
Abstract
Overall survival (OS)for glioblastoma multiforme (GBM) has a known association with the extent of tumor resection with gross total resection (GTR) typically considered as the upper limit. In certain regions such as the anterior temporal lobe, more extensive resection by means of a lobectomy may be feasible. In our systematic review and meta-analysis, we aimed to compare the outcomes of lobectomy and GTR for GBM. PubMed and Embase were queriedfor studies that compared the outcomes after lobectomy or GTR for GBM. The primary outcomes were OS, progression-free survival (PFS), and Karnofksy Performance Status (KPS) score at the latest follow-up. The secondary outcomes were seizure control at the latest follow-up and complication rates. Meta-analysis for OS and PFS was performed using individual-participant data reconstructed from published Kaplan-Meier curves. Random-effect meta-analysis was performed for KPS. The secondary outcomes were pooled using descriptive statistics. Of the 795 records screened, 6 were included in our study. Meta-analysis revealed that anterior temporal, frontal, or occipital lobectomy was associated with significantly better OS (p < 0.001) and PFS (p < 0.001) than GTR, but not KPS (MD = 6.37; 95% CI=(-13.80, 26.54); p = 0.536). Anterior temporal lobectomy was associated with significantly better seizure control rates than GTR for temporal GBM (OR = 27; 95% CI=(1.4, 515.9); p = 0.002). There was no statistically significant difference in complication rates between anterior temporal, frontal, or occipital lobectomy and GTR. In conclusion, lobectomy was associated with significantly better OS, PFS, and seizure control than GTR for GBM.
Collapse
Affiliation(s)
- Yilong Zheng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Seyed Ehsan Saffari
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore; Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - David Chyi Yeu Low
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Xuling Lin
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Justin Rui Xin Ker
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Samantha Ya Lyn Ang
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Wai Hoe Ng
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - Kai Rui Wan
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| |
Collapse
|
62
|
Cho NS, Wong WK, Nghiemphu PL, Cloughesy TF, Ellingson BM. The Future Glioblastoma Clinical Trials Landscape: Early Phase 0, Window of Opportunity, and Adaptive Phase I-III Studies. Curr Oncol Rep 2023; 25:1047-1055. [PMID: 37402043 PMCID: PMC10474988 DOI: 10.1007/s11912-023-01433-1] [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] [Accepted: 05/03/2023] [Indexed: 07/05/2023]
Abstract
PURPOSE OF REVIEW Innovative clinical trial designs for glioblastoma (GBM) are needed to expedite drug discovery. Phase 0, window of opportunity, and adaptive designs have been proposed, but their advanced methodologies and underlying biostatistics are not widely known. This review summarizes phase 0, window of opportunity, and adaptive phase I-III clinical trial designs in GBM tailored to physicians. RECENT FINDINGS Phase 0, window of opportunity, and adaptive trials are now being implemented for GBM. These trials can remove ineffective therapies earlier during drug development and improve trial efficiency. There are two ongoing adaptive platform trials: GBM Adaptive Global Innovative Learning Environment (GBM AGILE) and the INdividualized Screening trial of Innovative GBM Therapy (INSIGhT). The future clinical trials landscape in GBM will increasingly involve phase 0, window of opportunity, and adaptive phase I-III studies. Continued collaboration between physicians and biostatisticians will be critical for implementing these trial designs.
Collapse
Affiliation(s)
- Nicholas S Cho
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA, USA
- Medical Scientist Training Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Weng Kee Wong
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Phioanh L Nghiemphu
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Timothy F Cloughesy
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, 924 Westwood Blvd., Suite 615, Los Angeles, CA, 90024, USA.
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
63
|
Ortiz Rivera J, Velez Crespo G, Inyushin M, Kucheryavykh Y, Kucheryavykh L. Pyk2/FAK Signaling Is Upregulated in Recurrent Glioblastoma Tumors in a C57BL/6/GL261 Glioma Implantation Model. Int J Mol Sci 2023; 24:13467. [PMID: 37686276 PMCID: PMC10487692 DOI: 10.3390/ijms241713467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
The majority of glioblastomas (GBMs) recur shortly after tumor resection and recurrent tumors differ significantly from newly diagnosed GBMs, phenotypically and genetically. In this study, using a Gl261-C57Bl/6 mouse glioma implantation model, we identified significant upregulation of proline-rich tyrosine kinase Pyk2 and focal adhesion kinase (FAK) phosphorylation levels-pPyk2 (579/580) and pFAK (925)-without significant modifications in total Pyk2 and FAK protein expression in tumors regrown after surgical resection, compared with primary implanted tumors. Previously, we demonstrated that Pyk2 and FAK are involved in the regulation of tumor cell invasion and proliferation and are associated with reduced overall survival. We hypothesized that the use of inhibitors of Pyk2/FAK in the postsurgical period may reduce the growth of recurrent tumors. Using Western blot analysis and confocal immunofluorescence approaches, we demonstrated upregulation of Cyclin D1 and the Ki67 proliferation index in tumors regrown after resection, compared with primary implanted tumors. Treatment with Pyk2/FAK inhibitor PF-562271, administered through oral gavage at 50 mg/kg daily for two weeks beginning 2 days before tumor resection, reversed Pyk2/FAK signaling upregulation in recurrent tumors, reduced tumor volume, and increased animal survival. In conclusion, the use of Pyk2/FAK inhibitors can contribute to a delay in GBM tumor regrowth after surgical resection.
Collapse
Affiliation(s)
- Jescelica Ortiz Rivera
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956, USA; (G.V.C.); (M.I.); (Y.K.); (L.K.)
| | | | | | | | | |
Collapse
|
64
|
Liu X, Zhao Z, Dai W, Liao K, Sun Q, Chen D, Pan X, Feng L, Ding Y, Wei S. The Development of Immunotherapy for the Treatment of Recurrent Glioblastoma. Cancers (Basel) 2023; 15:4308. [PMID: 37686584 PMCID: PMC10486426 DOI: 10.3390/cancers15174308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/10/2023] Open
Abstract
Recurrent glioblastoma (rGBM) is a highly aggressive form of brain cancer that poses a significant challenge for treatment in neuro-oncology, and the survival status of patients after relapse usually means rapid deterioration, thus becoming the leading cause of death among patients. In recent years, immunotherapy has emerged as a promising strategy for the treatment of recurrent glioblastoma by stimulating the body's immune system to recognize and attack cancer cells, which could be used in combination with other treatments such as surgery, radiation, and chemotherapy to improve outcomes for patients with recurrent glioblastoma. This therapy combines several key methods such as the use of monoclonal antibodies, chimeric antigen receptor T cell (CAR-T) therapy, checkpoint inhibitors, oncolytic viral therapy cancer vaccines, and combination strategies. In this review, we mainly document the latest immunotherapies for the treatment of glioblastoma and especially focus on rGBM.
Collapse
Affiliation(s)
- Xudong Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Zihui Zhao
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Wufei Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering Research, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Kuo Liao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China;
| | - Qi Sun
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Dongjiang Chen
- Division of Neuro-Oncology, USC Keck Brain Tumor Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - Xingxin Pan
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Lishuang Feng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Ying Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Shiyou Wei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
65
|
Sousa N, Geiß C, Bindila L, Lieberwirth I, Kim E, Régnier-Vigouroux A. Targeting sphingolipid metabolism with the sphingosine kinase inhibitor SKI-II overcomes hypoxia-induced chemotherapy resistance in glioblastoma cells: effects on cell death, self-renewal, and invasion. BMC Cancer 2023; 23:762. [PMID: 37587449 PMCID: PMC10433583 DOI: 10.1186/s12885-023-11271-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: 10/12/2022] [Accepted: 08/07/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Glioblastoma patients commonly develop resistance to temozolomide chemotherapy. Hypoxia, which supports chemotherapy resistance, favors the expansion of glioblastoma stem cells (GSC), contributing to tumor relapse. Because of a deregulated sphingolipid metabolism, glioblastoma tissues contain high levels of the pro-survival sphingosine-1-phosphate and low levels of the pro-apoptotic ceramide. The latter can be metabolized to sphingosine-1-phosphate by sphingosine kinase (SK) 1 that is overexpressed in glioblastoma. The small molecule SKI-II inhibits SK and dihydroceramide desaturase 1, which converts dihydroceramide to ceramide. We previously reported that SKI-II combined with temozolomide induces caspase-dependent cell death, preceded by dihydrosphingolipids accumulation and autophagy in normoxia. In the present study, we investigated the effects of a low-dose combination of temozolomide and SKI-II under normoxia and hypoxia in glioblastoma cells and patient-derived GCSs. METHODS Drug synergism was analyzed with the Chou-Talalay Combination Index method. Dose-effect curves of each drug were determined with the Sulforhodamine B colorimetric assay. Cell death mechanisms and autophagy were analyzed by immunofluorescence, flow cytometry and western blot; sphingolipid metabolism alterations by mass spectrometry and gene expression analysis. GSCs self-renewal capacity was determined using extreme limiting dilution assays and invasion of glioblastoma cells using a 3D spheroid model. RESULTS Temozolomide resistance of glioblastoma cells was increased under hypoxia. However, combination of temozolomide (48 µM) with SKI-II (2.66 µM) synergistically inhibited glioblastoma cell growth and potentiated glioblastoma cell death relative to single treatments under hypoxia. This low-dose combination did not induce dihydrosphingolipids accumulation, but a decrease in ceramide and its metabolites. It induced oxidative and endoplasmic reticulum stress and triggered caspase-independent cell death. It impaired the self-renewal capacity of temozolomide-resistant GSCs, especially under hypoxia. Furthermore, it decreased invasion of glioblastoma cell spheroids. CONCLUSIONS This in vitro study provides novel insights on the links between sphingolipid metabolism and invasion, a hallmark of cancer, and cancer stem cells, key drivers of cancer. It demonstrates the therapeutic potential of approaches that combine modulation of sphingolipid metabolism with first-line agent temozolomide in overcoming tumor growth and relapse by reducing hypoxia-induced resistance to chemotherapy and by targeting both differentiated and stem glioblastoma cells.
Collapse
Affiliation(s)
- Nadia Sousa
- Institute of Developmental Biology & Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Carsten Geiß
- Institute of Developmental Biology & Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Laura Bindila
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, Medical University Mainz, Mainz, Germany
| | | | - Ella Kim
- Department of Neurosurgery, Medical University of Mainz, Mainz, Germany
| | - Anne Régnier-Vigouroux
- Institute of Developmental Biology & Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
66
|
Choi SW, Jung HA, Cho H, Kim TM, Park C, Nam D, Lee S. A multicenter, phase II trial of GC1118, a novel anti-EGFR antibody, for recurrent glioblastoma patients with EGFR amplification. Cancer Med 2023; 12:15788-15796. [PMID: 37537946 PMCID: PMC10469652 DOI: 10.1002/cam4.6213] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND We evaluated the therapeutic efficacy of GC1118, a novel anti-epidermal growth factor receptor (EGFR) monoclonal antibody, in recurrent glioblastoma (GBM) patients with EGFR amplification. METHODS This study was a multicenter, open-label, single-arm phase II trial. Recurrent GBM patients with EGFR amplification were eligible: EGFR amplification was determined using fluorescence in situ hybridization analysis when a sample had both the EGFR/CEP7 ratio of ≥2 and a tight cluster EGFR signal in ≥10% of recorded cells. GC1118 was administered intravenously at a dose of 4 mg/kg once weekly. The primary endpoint was the 6-month progression-free survival rate (PFS6). Next-generation sequencing was performed to investigate the molecular biomarkers related to the response to GC1118. RESULTS Between April 2018 and December 2020, 21 patients were enrolled in the study and received GC1118 treatment. Eighteen patients were eligible for efficacy analysis. The PFS6 was 5.6% (95% confidence interval, 0.3%-25.8%, Wilson method). The median progression-free survival was 1.7 months (range: 28 days-7.2 months) and median overall survival was 5.7 months (range: 2-22.0 months). GC1118 was well tolerated except skin toxicities. Skin rash was the most frequent adverse event and four patients experienced Grade 3 skin-related toxicity. Genomic analysis revealed that the immune-related signatures were upregulated in patients with tumor regression. CONCLUSION This study did not meet the primary endpoint (PFS6); however, we found that immune signatures were significantly upregulated in the tumors with regression upon GC1118 therapy, which signifies the potential of immune-mediated antitumor efficacy of GC1118.
Collapse
Affiliation(s)
- Seung Won Choi
- Department of NeurosurgerySchool of Medicine, Sungkyunkwan University, Samsung Medical CenterSeoulRepublic of Korea
- Present address:
Program for Mathematical Genomics and Department of Systems BiologyColumbia UniversityNew YorkNYUSA
| | - Hyun Ae Jung
- Department of Medicine, Division of Hematology‐OncologySchool of Medicine, Sungkyunkwan University, Samsung Medical CenterSeoulRepublic of Korea
| | - Hee‐Jin Cho
- Department of Biomedical Convergence Science and TechnologyKyungpook National UniversityDaeguRepublic of Korea
| | - Tae Min Kim
- Department of Internal MedicineSeoul National University Hospital, Seoul National University Cancer Research Institute, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Chul‐Kee Park
- Department of NeurosurgerySeoul National University Hospital, College of MedicineSeoulRepublic of Korea
| | - Do‐Hyun Nam
- Department of NeurosurgerySchool of Medicine, Sungkyunkwan University, Samsung Medical CenterSeoulRepublic of Korea
| | - Se‐Hoon Lee
- Department of Medicine, Division of Hematology‐OncologySchool of Medicine, Sungkyunkwan University, Samsung Medical CenterSeoulRepublic of Korea
| |
Collapse
|
67
|
Mantica M, Drappatz J, Lieberman F, Hadjipanayis CG, Lunsford LD, Niranjan A. Phase II study of border zone stereotactic radiosurgery with bevacizumab in patients with recurrent or progressive glioblastoma multiforme. J Neurooncol 2023; 164:179-190. [PMID: 37515669 DOI: 10.1007/s11060-023-04398-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/14/2023] [Indexed: 07/31/2023]
Abstract
PURPOSE Recurrent glioblastoma is universally fatal with limited effective treatment options. The aim of this phase 2 study of Border Zone SRS plus bevacizumab was to evaluate OS in patients with recurrent GBM. METHODS Patients with histologically confirmed GBM with recurrent disease who had received prior first-line treatment with fractionated radiotherapy and chemotherapy and eligible for SRS were enrolled. Bevacizumab 10 mg/kg was given day -1, day 14, and then every 14 days until disease progression. 1-14 days before BZ-SRS procedure, patients underwent brain MRI /MRS. MRS with measurement of choline-to-N-acetyl aspartate index (CNI) area ≥ 3 was targeted for SRS. RESULTS From 2015-2017, sixteen of planned 40 patients were enrolled. The median age was 62 (range, 48-74Y). 3/16 (0.188) participants experienced grade 2 toxicity. No AREs were reported. The mOS was 11.73 months compared to 8.74 months (P = 0.324) from date of SRS for the BZ-SRS and institutional historical controls, respectively. PFS-6 and OS-6 were 31.2% (p = 0.00294) and 81.2%(p = 0.058), respectively. Of 13 evaluable for best response: 1 CR (p = 0.077), 4 PR (p = 0.308), 7 SD (p = 0.538), and 1 PD (p = 0.077). 11/16 participants had MRS scans with an estimated probability that MRS changes a treatment plan of 0 (0, 0.285). CONCLUSION BZ-SRS with bevacizumab was feasible and well tolerated. There is no significant survival benefit using BZ-SRS with bevacizumab compared to institutional historical controls. Secondary analysis revealed a trend toward improved PFS-6, but not OS-6 after BZ-SRS. MRS scans did not result in changes to SRS treatment plans.
Collapse
Affiliation(s)
- Megan Mantica
- University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA.
| | - Jan Drappatz
- University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA
| | - Frank Lieberman
- University of Pittsburgh Medical Center, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA
| | | | - L Dade Lunsford
- University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Ajay Niranjan
- University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| |
Collapse
|
68
|
Hsia T, Small JL, Yekula A, Batool SM, Escobedo AK, Ekanayake E, You DG, Lee H, Carter BS, Balaj L. Systematic Review of Photodynamic Therapy in Gliomas. Cancers (Basel) 2023; 15:3918. [PMID: 37568734 PMCID: PMC10417382 DOI: 10.3390/cancers15153918] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Over the last 20 years, gliomas have made up over 89% of malignant CNS tumor cases in the American population (NIH SEER). Within this, glioblastoma is the most common subtype, comprising 57% of all glioma cases. Being highly aggressive, this deadly disease is known for its high genetic and phenotypic heterogeneity, rendering a complicated disease course. The current standard of care consists of maximally safe tumor resection concurrent with chemoradiotherapy. However, despite advances in technology and therapeutic modalities, rates of disease recurrence are still high and survivability remains low. Given the delicate nature of the tumor location, remaining margins following resection often initiate disease recurrence. Photodynamic therapy (PDT) is a therapeutic modality that, following the administration of a non-toxic photosensitizer, induces tumor-specific anti-cancer effects after localized, wavelength-specific illumination. Its effect against malignant glioma has been studied extensively over the last 30 years, in pre-clinical and clinical trials. Here, we provide a comprehensive review of the three generations of photosensitizers alongside their mechanisms of action, limitations, and future directions.
Collapse
Affiliation(s)
- Tiffaney Hsia
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Julia L. Small
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Chan Medical School, University of Massachusetts, Worcester, MA 01605, USA
| | - Anudeep Yekula
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 554414, USA
| | - Syeda M. Batool
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ana K. Escobedo
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Emil Ekanayake
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Dong Gil You
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA 02114, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
69
|
Martucci M, Russo R, Giordano C, Schiarelli C, D’Apolito G, Tuzza L, Lisi F, Ferrara G, Schimperna F, Vassalli S, Calandrelli R, Gaudino S. Advanced Magnetic Resonance Imaging in the Evaluation of Treated Glioblastoma: A Pictorial Essay. Cancers (Basel) 2023; 15:3790. [PMID: 37568606 PMCID: PMC10417432 DOI: 10.3390/cancers15153790] [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: 06/15/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
MRI plays a key role in the evaluation of post-treatment changes, both in the immediate post-operative period and during follow-up. There are many different treatment's lines and many different neuroradiological findings according to the treatment chosen and the clinical timepoint at which MRI is performed. Structural MRI is often insufficient to correctly interpret and define treatment-related changes. For that, advanced MRI modalities, including perfusion and permeability imaging, diffusion tensor imaging, and magnetic resonance spectroscopy, are increasingly utilized in clinical practice to characterize treatment effects more comprehensively. This article aims to provide an overview of the role of advanced MRI modalities in the evaluation of treated glioblastomas. For a didactic purpose, we choose to divide the treatment history in three main timepoints: post-surgery, during Stupp (first-line treatment) and at recurrence (second-line treatment). For each, a brief introduction, a temporal subdivision (when useful) or a specific drug-related paragraph were provided. Finally, the current trends and application of radiomics and artificial intelligence (AI) in the evaluation of treated GB have been outlined.
Collapse
Affiliation(s)
- Matia Martucci
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, 00168 Rome, Italy; (R.R.); (C.G.); (C.S.); (G.D.); (R.C.); (S.G.)
| | - Rosellina Russo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, 00168 Rome, Italy; (R.R.); (C.G.); (C.S.); (G.D.); (R.C.); (S.G.)
| | - Carolina Giordano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, 00168 Rome, Italy; (R.R.); (C.G.); (C.S.); (G.D.); (R.C.); (S.G.)
| | - Chiara Schiarelli
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, 00168 Rome, Italy; (R.R.); (C.G.); (C.S.); (G.D.); (R.C.); (S.G.)
| | - Gabriella D’Apolito
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, 00168 Rome, Italy; (R.R.); (C.G.); (C.S.); (G.D.); (R.C.); (S.G.)
| | - Laura Tuzza
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.T.); (F.L.); (G.F.); (F.S.); (S.V.)
| | - Francesca Lisi
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.T.); (F.L.); (G.F.); (F.S.); (S.V.)
| | - Giuseppe Ferrara
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.T.); (F.L.); (G.F.); (F.S.); (S.V.)
| | - Francesco Schimperna
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.T.); (F.L.); (G.F.); (F.S.); (S.V.)
| | - Stefania Vassalli
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.T.); (F.L.); (G.F.); (F.S.); (S.V.)
| | - Rosalinda Calandrelli
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, 00168 Rome, Italy; (R.R.); (C.G.); (C.S.); (G.D.); (R.C.); (S.G.)
| | - Simona Gaudino
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico “A. Gemelli” IRCCS, 00168 Rome, Italy; (R.R.); (C.G.); (C.S.); (G.D.); (R.C.); (S.G.)
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.T.); (F.L.); (G.F.); (F.S.); (S.V.)
| |
Collapse
|
70
|
Andrieux G, Das T, Griffin M, Straehle J, Paine SML, Beck J, Boerries M, Heiland DH, Smith SJ, Rahman R, Chakraborty S. Spatially resolved transcriptomic profiles reveal unique defining molecular features of infiltrative 5ALA-metabolizing cells associated with glioblastoma recurrence. Genome Med 2023; 15:48. [PMID: 37434262 PMCID: PMC10337060 DOI: 10.1186/s13073-023-01207-1] [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: 07/01/2022] [Accepted: 06/26/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Spatiotemporal heterogeneity originating from genomic and transcriptional variation was found to contribute to subtype switching in isocitrate dehydrogenase-1 wild-type glioblastoma (GBM) prior to and upon recurrence. Fluorescence-guided neurosurgical resection utilizing 5-aminolevulinic acid (5ALA) enables intraoperative visualization of infiltrative tumors outside the magnetic resonance imaging contrast-enhanced regions. The cell population and functional status of tumor responsible for enhancing 5ALA-metabolism to fluorescence-active PpIX remain elusive. The close spatial proximity of 5ALA-metabolizing (5ALA +) cells to residual disease remaining post-surgery renders 5ALA + biology an early a priori proxy of GBM recurrence, which is poorly understood. METHODS We performed spatially resolved bulk RNA profiling (SPRP) analysis of unsorted Core, Rim, Invasive margin tissue, and FACS-isolated 5ALA + /5ALA - cells from the invasive margin across IDH-wt GBM patients (N = 10) coupled with histological, radiographic, and two-photon excitation fluorescence microscopic analyses. Deconvolution of SPRP followed by functional analyses was performed using CIBEROSRTx and UCell enrichment algorithms, respectively. We further investigated the spatial architecture of 5ALA + enriched regions by analyzing spatial transcriptomics from an independent IDH-wt GBM cohort (N = 16). Lastly, we performed survival analysis using Cox Proportinal-Hazards model on large GBM cohorts. RESULTS SPRP analysis integrated with single-cell and spatial transcriptomics uncovered that the GBM molecular subtype heterogeneity is likely to manifest regionally in a cell-type-specific manner. Infiltrative 5ALA + cell population(s) harboring transcriptionally concordant GBM and myeloid cells with mesenchymal subtype, -active wound response, and glycolytic metabolic signature, was shown to reside within the invasive margin spatially distinct from the tumor core. The spatial co-localization of the infiltrating MES GBM and myeloid cells within the 5ALA + region indicates PpIX fluorescence can effectively be utilized to resect the immune reactive zone beyond the tumor core. Finally, 5ALA + gene signatures were associated with poor survival and recurrence in GBM, signifying that the transition from primary to recurrent GBM is not discrete but rather a continuum whereby primary infiltrative 5ALA + remnant tumor cells more closely resemble the eventual recurrent GBM. CONCLUSIONS Elucidating the unique molecular and cellular features of the 5ALA + population within tumor invasive margin opens up unique possibilities to develop more effective treatments to delay or block GBM recurrence, and warrants commencement of such treatments as early as possible post-surgical resection of the primary neoplasm.
Collapse
Affiliation(s)
- Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tonmoy Das
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Systems Cell-Signaling Laboratory, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Michaela Griffin
- Children's Brain Tumour Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jakob Straehle
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Simon M L Paine
- Children's Brain Tumour Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jürgen Beck
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dieter H Heiland
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Microenvironment and Immunology Research Laboratory, Medical Center - University of Freiburg, Freiburg, Germany
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Stuart J Smith
- Children's Brain Tumour Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ruman Rahman
- Children's Brain Tumour Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK.
| | - Sajib Chakraborty
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Systems Cell-Signaling Laboratory, Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh.
| |
Collapse
|
71
|
Moreno V, Manuel Sepúlveda J, Reardon DA, Pérez-Núñez Á, González León P, Hanna B, Filvaroff E, Aronchik I, Chang H, Amoroso B, Zuraek M, Sanchez-Perez T, Mendez C, Stephens D, Nikolova Z, Vogelbaum MA. Trotabresib, an oral potent bromodomain and extraterminal inhibitor, in patients with high-grade gliomas: A phase I, "window-of-opportunity" study. Neuro Oncol 2023; 25:1113-1122. [PMID: 36455228 PMCID: PMC10237409 DOI: 10.1093/neuonc/noac263] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND The bromodomain and extraterminal protein (BET) inhibitor trotabresib has demonstrated antitumor activity in patients with advanced solid tumors, including high-grade gliomas. CC-90010-GBM-001 (NCT04047303) is a phase I study investigating the pharmacokinetics, pharmacodynamics, and CNS penetration of trotabresib in patients with recurrent high-grade gliomas scheduled for salvage resection. METHODS Patients received trotabresib 30 mg/day on days 1-4 before surgery, followed by maintenance trotabresib 45 mg/day 4 days on/24 days off after surgery. Primary endpoints were plasma pharmacokinetics and trotabresib concentrations in resected tissue. Secondary and exploratory endpoints included safety, pharmacodynamics, and antitumor activity. RESULTS Twenty patients received preoperative trotabresib and underwent resection with no delays or cancelations of surgery; 16 patients received maintenance trotabresib after recovery from surgery. Trotabresib plasma pharmacokinetics were consistent with previous data. Mean trotabresib brain tumor tissue:plasma ratio was 0.84 (estimated unbound partition coefficient [KPUU] 0.37), and modulation of pharmacodynamic markers was observed in blood and brain tumor tissue. Trotabresib was well tolerated; the most frequent grade 3/4 treatment-related adverse event during maintenance treatment was thrombocytopenia (5/16 patients). Six-month progression-free survival was 12%. Two patients remain on treatment with stable disease at cycles 25 and 30. CONCLUSIONS Trotabresib penetrates the blood-brain-tumor barrier in patients with recurrent high-grade glioma and demonstrates target engagement in resected tumor tissue. Plasma pharmacokinetics, blood pharmacodynamics, and safety were comparable with previous results for trotabresib in patients with advanced solid tumors. Investigation of adjuvant trotabresib + temozolomide and concomitant trotabresib + temozolomide + radiotherapy in patients with newly diagnosed glioblastoma is ongoing (NCT04324840).
Collapse
Affiliation(s)
- Victor Moreno
- START Madrid-FJD, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | | | - David A Reardon
- Department of Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ángel Pérez-Núñez
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Pedro González León
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Bishoy Hanna
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Ida Aronchik
- Bristol Myers Squibb, San Francisco, California, USA
| | - Henry Chang
- Bristol Myers Squibb, San Francisco, California, USA
| | - Barbara Amoroso
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | | | - Tania Sanchez-Perez
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | - Cristina Mendez
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | | | - Zariana Nikolova
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | | |
Collapse
|
72
|
Sampson JH, Singh Achrol A, Aghi MK, Bankiewicz K, Bexon M, Brem S, Brenner A, Chandhasin C, Chowdhary S, Coello M, Ellingson BM, Floyd JR, Han S, Kesari S, Mardor Y, Merchant F, Merchant N, Randazzo D, Vogelbaum M, Vrionis F, Wembacher-Schroeder E, Zabek M, Butowski N. Targeting the IL4 receptor with MDNA55 in patients with recurrent glioblastoma: Results of a phase IIb trial. Neuro Oncol 2023; 25:1085-1097. [PMID: 36640127 PMCID: PMC10237418 DOI: 10.1093/neuonc/noac285] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND MDNA55 is an interleukin 4 receptor (IL4R)-targeting toxin in development for recurrent GBM, a universally fatal disease. IL4R is overexpressed in GBM as well as cells of the tumor microenvironment. High expression of IL4R is associated with poor clinical outcomes. METHODS MDNA55-05 is an open-label, single-arm phase IIb study of MDNA55 in recurrent GBM (rGBM) patients with an aggressive form of GBM (de novo GBM, IDH wild-type, and nonresectable at recurrence) on their 1st or 2nd recurrence. MDNA55 was administered intratumorally as a single dose treatment (dose range of 18 to 240 ug) using convection-enhanced delivery (CED) with up to 4 stereo-tactically placed catheters. It was co-infused with a contrast agent (Gd-DTPA, Magnevist®) to assess distribution in and around the tumor margins. The flow rate of each catheter did not exceed 10μL/min to ensure that the infusion duration did not exceed 48 h. The primary endpoint was mOS, with secondary endpoints determining the effects of IL4R status on mOS and PFS. RESULTS MDNA55 showed an acceptable safety profile at doses up to 240 μg. In all evaluable patients (n = 44) mOS was 11.64 months (80% one-sided CI 8.62, 15.02) and OS-12 was 46%. A subgroup (n = 32) consisting of IL4R High and IL4R Low patients treated with high-dose MDNA55 (>180 ug) showed the best benefit with mOS of 15 months, OS-12 of 55%. Based on mRANO criteria, tumor control was observed in 81% (26/32), including those patients who exhibited pseudo-progression (15/26). CONCLUSIONS MDNA55 demonstrated tumor control and promising survival and may benefit rGBM patients when treated at high-dose irrespective of IL4R expression level.Trial Registration: Clinicaltrials.gov NCT02858895.
Collapse
Affiliation(s)
- John H Sampson
- Duke University Medical Center, Department of Neurosurgery, Durham, North Carolina, USA
| | - Achal Singh Achrol
- Loma Linda University Medical Center, Department of Neurosurgery, Loma Linda, California, USA
| | - Manish K Aghi
- University of California San Francisco, Department of Neurological Surgery, San Francisco, California, USA
| | - Krystof Bankiewicz
- Ohio State University College of Medicine, Department of Neurological Surgery, Columbus, Ohio, USA
| | | | - Steven Brem
- Hospital of the University of Pennsylvania, Department of Neurosurgery, Philadelphia, Pennsylvania, USA
| | - Andrew Brenner
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | | | | | | | - Benjamin M Ellingson
- University of California, Los Angeles, Brain Tumor Imaging Laboratory (BTIL), California, USA
| | - John R Floyd
- University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Seunggu Han
- Oregon Health & Science University, Portland, Oregon, USA
| | - Santosh Kesari
- Pacific Neurosciences Institute, Santa Monica, California, USA
| | | | | | | | - Dina Randazzo
- Duke University Medical Center, Department of Neurosurgery, Durham, North Carolina, USA
| | - Michael Vogelbaum
- H. Lee Moffitt Cancer Center & Research Institute, Department of Neuro-Oncology, Tampa, Florida, USA
| | - Frank Vrionis
- Boca Raton Regional Hospital, Boca Raton, Florida, USA
| | | | | | - Nicholas Butowski
- University of California San Francisco, Department of Neurological Surgery, San Francisco, California, USA
| |
Collapse
|
73
|
Young D, Guha C, Sidoli S. The role of histone H3 lysine demethylases in glioblastoma. Cancer Metastasis Rev 2023; 42:445-454. [PMID: 37286866 DOI: 10.1007/s10555-023-10114-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023]
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults with an average survival of 15-18 months. Part of its malignancy derives from epigenetic regulation that occurs as the tumor develops and after therapeutic treatment. Specifically, enzymes involved in removing methylations from histone proteins on chromatin, i.e., lysine demethylases (KDMs), have a significant impact on GBM biology and reoccurrence. This knowledge has paved the way to considering KDMs as potential targets for GBM treatment. For example, increases in trimethylation of histone H3 on the lysine 9 residue (H3K9me3) via inhibition of KDM4C and KDM7A has been shown to lead to cell death in Glioblastoma initiating cells. KDM6 has been shown to drive Glioma resistance to receptor tyrosine kinase inhibitors and its inhibition decreases tumor resistance. In addition, increased expression of the histone methyltransferase MLL4 and UTX histone demethylase are associated with prolonged survival in a subset of GBM patients, potentially by regulating histone methylation on the promoter of the mgmt gene. Thus, the complexity of how histone modifiers contribute to glioblastoma pathology and disease progression is yet to be fully understood. To date, most of the current work on histone modifying enzymes in GBM are centered upon histone H3 demethylase enzymes. In this mini-review, we summarize the current knowledge on the role of histone H3 demethylase enzymes in Glioblastoma tumor biology and therapy resistance. The objective of this work is to highlight the current and future potential areas of research for GBM epigenetics therapy.
Collapse
Affiliation(s)
- Dejauwne Young
- Department of Biochemistry, Albert Einstein College of Medicine, The Bronx, New York City, NY, 10461, USA
- Department of Radiation Oncology, Department of Pathology, Department of Urology, Albert Einstein College of Medicine, The Bronx, New York City, NY, 10461, USA
| | - Chandan Guha
- Department of Radiation Oncology, Department of Pathology, Department of Urology, Albert Einstein College of Medicine, The Bronx, New York City, NY, 10461, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, The Bronx, New York City, NY, 10461, USA.
| |
Collapse
|
74
|
Higginbottom SL, Tomaskovic-Crook E, Crook JM. Considerations for modelling diffuse high-grade gliomas and developing clinically relevant therapies. Cancer Metastasis Rev 2023; 42:507-541. [PMID: 37004686 PMCID: PMC10348989 DOI: 10.1007/s10555-023-10100-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/16/2023] [Indexed: 04/04/2023]
Abstract
Diffuse high-grade gliomas contain some of the most dangerous human cancers that lack curative treatment options. The recent molecular stratification of gliomas by the World Health Organisation in 2021 is expected to improve outcomes for patients in neuro-oncology through the development of treatments targeted to specific tumour types. Despite this promise, research is hindered by the lack of preclinical modelling platforms capable of recapitulating the heterogeneity and cellular phenotypes of tumours residing in their native human brain microenvironment. The microenvironment provides cues to subsets of glioma cells that influence proliferation, survival, and gene expression, thus altering susceptibility to therapeutic intervention. As such, conventional in vitro cellular models poorly reflect the varied responses to chemotherapy and radiotherapy seen in these diverse cellular states that differ in transcriptional profile and differentiation status. In an effort to improve the relevance of traditional modelling platforms, recent attention has focused on human pluripotent stem cell-based and tissue engineering techniques, such as three-dimensional (3D) bioprinting and microfluidic devices. The proper application of these exciting new technologies with consideration of tumour heterogeneity and microenvironmental interactions holds potential to develop more applicable models and clinically relevant therapies. In doing so, we will have a better chance of translating preclinical research findings to patient populations, thereby addressing the current derisory oncology clinical trial success rate.
Collapse
Affiliation(s)
- Sarah L Higginbottom
- Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Fairy Meadow, NSW, 2519, Australia
- Arto Hardy Family Biomedical Innovation Hub, Chris O'Brien Lifehouse, Camperdown, NSW, 2050, Australia
| | - Eva Tomaskovic-Crook
- Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Fairy Meadow, NSW, 2519, Australia.
- Arto Hardy Family Biomedical Innovation Hub, Chris O'Brien Lifehouse, Camperdown, NSW, 2050, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Jeremy M Crook
- Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Fairy Meadow, NSW, 2519, Australia.
- Arto Hardy Family Biomedical Innovation Hub, Chris O'Brien Lifehouse, Camperdown, NSW, 2050, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
75
|
Boulhen C, AIT SSI S, Benthami H, Razzouki I, Lakhdar A, Karkouri M, Badou A. TMIGD2 as a potential therapeutic target in glioma patients. Front Immunol 2023; 14:1173518. [PMID: 37261362 PMCID: PMC10227580 DOI: 10.3389/fimmu.2023.1173518] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/21/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction Among all types of central nervous system cancers, glioma remains the most frequent primary brain tumor in adults. Despite significant advances in immunomodulatory therapies, notably immune checkpoint inhibitors, their effectiveness remains constrained due to glioma resistance. The discovery of TMIGD2 (Transmembrane and Immunoglobulin Domain Containing 2) as an immuno-stimulatory receptor, constitutively expressed on naive T cells and most natural killer (NK) cells, has emerged as an attractive immunotherapy target in a variety of cancers. The expression profile of TMIGD2 and its significance in the overall survival of glioma patients remains unknown. Methods In the present study, we first assessed TMIGD2 mRNA expression using the Cancer Genome Atlas (TCGA) glioma transcriptome dataset (667 patients), followed by validation with the Chinese Glioma Genome Atlas (CGGA) cohort (693 patients). Secondly, we examined TMIGD2 protein staining in a series of 25 paraffin-embedded blocks from Moroccan glioma patients. The statistical analysis was performed using GraphPad Prism 8 software. Results TMIGD2 expression was found to be significantly higher in astrocytoma, IDH-1 mutations, low-grade, and young glioma patients. TMIGD2 was expressed on immune cells and, surprisingly, on tumor cells of glioma patients. Interestingly, our study demonstrated that TMIGD2 expression was negatively correlated with angiogenesis, hypoxia, G2/M checkpoint, and epithelial to mesenchymal transition signaling pathways. We also demonstrated that dendritic cells, monocytes, NK cells, gd T cells, and naive CD8 T cell infiltration correlates positively with TMIGD2 expression. On the other hand, Mantel-Cox analysis demonstrated that increased expression of TMIGD2 in human gliomas is associated with good overall survival. Cox multivariable analysis revealed that TMIGD2 is an independent predictor of a good prognosis in glioma patients. Discussion Taken together, our results highlight the tight implication of TMIGD2 in glioma progression and show its promising therapeutic potential as a stimulatory target for immunotherapy.
Collapse
Affiliation(s)
- Chaimae Boulhen
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Saadia AIT SSI
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Hamza Benthami
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Ibtissam Razzouki
- Laboratory of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd, Hassan II University, Casablanca, Morocco
| | - Abdelhakim Lakhdar
- Department of Neurosurgery, Faculty of Medicine and Pharmacy, University of Hassan II, Casablanca, Morocco
| | - Mehdi Karkouri
- Laboratory of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd, Hassan II University, Casablanca, Morocco
| | - Abdallah Badou
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco and Mohammed VI University of Sciences and Health, Casablanca, Morocco
| |
Collapse
|
76
|
Ranjan T, Sengupta S, Glantz MJ, Green RM, Yu A, Aregawi D, Chaudhary R, Chen R, Zuccarello M, Lu-Emerson C, Moulding HD, Belman N, Glass J, Mammoser A, Anderson M, Valluri J, Marko N, Schroeder J, Jubelirer S, Chow F, Claudio PP, Alberico AM, Lirette ST, Denning KL, Howard CM. Cancer stem cell assay-guided chemotherapy improves survival of patients with recurrent glioblastoma in a randomized trial. Cell Rep Med 2023; 4:101025. [PMID: 37137304 PMCID: PMC10213810 DOI: 10.1016/j.xcrm.2023.101025] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/19/2022] [Accepted: 04/10/2023] [Indexed: 05/05/2023]
Abstract
Therapy-resistant cancer stem cells (CSCs) contribute to the poor clinical outcomes of patients with recurrent glioblastoma (rGBM) who fail standard of care (SOC) therapy. ChemoID is a clinically validated assay for identifying CSC-targeted cytotoxic therapies in solid tumors. In a randomized clinical trial (NCT03632135), the ChemoID assay, a personalized approach for selecting the most effective treatment from FDA-approved chemotherapies, improves the survival of patients with rGBM (2016 WHO classification) over physician-chosen chemotherapy. In the ChemoID assay-guided group, median survival is 12.5 months (95% confidence interval [CI], 10.2-14.7) compared with 9 months (95% CI, 4.2-13.8) in the physician-choice group (p = 0.010) as per interim efficacy analysis. The ChemoID assay-guided group has a significantly lower risk of death (hazard ratio [HR] = 0.44; 95% CI, 0.24-0.81; p = 0.008). Results of this study offer a promising way to provide more affordable treatment for patients with rGBM in lower socioeconomic groups in the US and around the world.
Collapse
Affiliation(s)
- Tulika Ranjan
- Department of Neuro-Oncology, Allegheny Health Network, Pittsburgh, PA, USA; Department of Neuro-Oncology, Cancer Center Southern Florida, Tampa General Hospital, Tampa, FL, USA
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Michael J Glantz
- Department of Neurosurgery, Penn State Neuroscience Institute, Hershey, PA, USA
| | - Richard M Green
- Department of Neuro-Oncology, Southern California Permanente Medical Group, Los Angeles, CA, USA
| | - Alexander Yu
- Department of Neurosurgery, Allegheny Health Network, Pittsburgh, PA, USA
| | - Dawit Aregawi
- Department of Neurosurgery, Penn State Neuroscience Institute, Hershey, PA, USA
| | - Rekha Chaudhary
- Department of Internal Medicine, Division of Hematology-Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Ricky Chen
- Department of Neuro-Oncology, Providence Brain & Spine Institute, Portland, OR, USA
| | - Mario Zuccarello
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, USA
| | | | - Hugh D Moulding
- Department of Neuroscience, St. Luke's University Hospital & Health Network, Bethlehem, PA, USA
| | - Neil Belman
- Department of Neuroscience, St. Luke's University Hospital & Health Network, Bethlehem, PA, USA
| | - Jon Glass
- Departments of Neurology and Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Aaron Mammoser
- Department of Neurosurgery, LSU Health Sciences Center, New Orleans, LA, USA
| | - Mark Anderson
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Nicholas Marko
- Department of Neurosurgery, LewisGale Regional Health System, Salem, VA, USA
| | - Jason Schroeder
- Department of Neurosurgery, University of Toledo, Toledo, OH, USA
| | - Steven Jubelirer
- Department of Neuro-Oncology, Charleston Area Medical Center, Charleston, WV, USA
| | - Frances Chow
- Departments of Neurological Surgery and Neurology, University of Southern California, Los Angeles, CA, USA
| | - Pier Paolo Claudio
- Cordgenics, LLC, Huntington WV, USA; Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Anthony M Alberico
- Department of Neurosurgery, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Seth T Lirette
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Krista L Denning
- Department of Pathology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Candace M Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
77
|
Muller Bark J, Karpe AV, Doecke JD, Leo P, Jeffree RL, Chua B, Day BW, Beale DJ, Punyadeera C. A pilot study: Metabolic profiling of plasma and saliva samples from newly diagnosed glioblastoma patients. Cancer Med 2023; 12:11427-11437. [PMID: 37031458 PMCID: PMC10242862 DOI: 10.1002/cam4.5857] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/04/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
BACKGROUND Despite aggressive treatment, more than 90% of glioblastoma (GBM) patients experience recurrences. GBM response to therapy is currently assessed by imaging techniques and tissue biopsy. However, difficulties with these methods may cause misinterpretation of treatment outcomes. Currently, no validated therapy response biomarkers are available for monitoring GBM progression. Metabolomics holds potential as a complementary tool to improve the interpretation of therapy responses to help in clinical interventions for GBM patients. METHODS Saliva and blood from GBM patients were collected pre and postoperatively. Patients were stratified conforming their progression-free survival (PFS) into favourable or unfavourable clinical outcomes (>9 months or PFS ≤ 9 months, respectively). Analysis of saliva (whole-mouth and oral rinse) and plasma samples was conducted utilising LC-QqQ-MS and LC-QTOF-MS to determine the metabolomic and lipidomic profiles. The data were investigated using univariate and multivariate statistical analyses and graphical LASSO-based graphic network analyses. RESULTS Altogether, 151 metabolites and 197 lipids were detected within all saliva and plasma samples. Among the patients with unfavourable outcomes, metabolites such as cyclic-AMP, 3-hydroxy-kynurenine, dihydroorotate, UDP and cis-aconitate were elevated, compared to patients with favourable outcomes during pre-and post-surgery. These metabolites showed to impact the pentose phosphate and Warburg effect pathways. The lipid profile of patients who experienced unfavourable outcomes revealed a higher heterogeneity in the abundance of lipids and fewer associations between markers in contrast to the favourable outcome group. CONCLUSION Our findings indicate that changes in salivary and plasma metabolites in GBM patients can potentially be employed as less invasive prognostic biomarkers/biomarker panel but validation with larger cohorts is required.
Collapse
Affiliation(s)
- Juliana Muller Bark
- Faculty of Health, Centre for Biomedical TechnologiesSchool of Biomedical Sciences, Queensland University of TechnologyBrisbaneQueenslandAustralia
- Saliva and Liquid Biopsy Translational LaboratoryGriffith Institute for Drug Discovery – Griffith UniversityBrisbaneQueenslandAustralia
- Faculty of HealthSchool of Biomedical Sciences, Queensland University of TechnologyGardens PointQueenslandAustralia
| | - Avinash V. Karpe
- Environment, Commonwealth Scientific and Industrial Research Organization (CSIRO), Ecosciences PrecinctDutton ParkQueenslandAustralia
- Agriculture and FoodCommonwealth Scientific and Industrial Research Organization (CSIRO)ActonAustralian Capital TerritoryAustralia
| | - James D. Doecke
- Australian eHealth Research Centre, CSIRO. Level 7, Surgical Treatment and Rehabilitation Service – STARSRoyal Brisbane and Women's HospitalHerstonQueenslandAustralia
| | - Paul Leo
- Faculty of HealthSchool of Biomedical Sciences, Queensland University of TechnologyGardens PointQueenslandAustralia
- Faculty of Health, Translational Genomics GroupSchool of Biomedical Sciences, Queensland University of TechnologyWoolloongabbaAustralia
| | - Rosalind L. Jeffree
- QIMR Berghofer Medical Research InstituteHerstonQueenslandAustralia
- Faculty of MedicineUniversity of QueenslandHerstonQueenslandAustralia
- Kenneth G. Jamieson Department of NeurosurgeryRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
- Cell and Molecular Biology Department, Sid Faithfull Brain Cancer LaboratoryQIMR Berghofer MRIBrisbaneQueenslandAustralia
| | - Benjamin Chua
- Faculty of MedicineUniversity of QueenslandHerstonQueenslandAustralia
- Cancer Care ServicesRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Bryan W. Day
- Faculty of HealthSchool of Biomedical Sciences, Queensland University of TechnologyGardens PointQueenslandAustralia
- Faculty of MedicineUniversity of QueenslandHerstonQueenslandAustralia
- Cell and Molecular Biology Department, Sid Faithfull Brain Cancer LaboratoryQIMR Berghofer MRIBrisbaneQueenslandAustralia
| | - David J. Beale
- Environment, Commonwealth Scientific and Industrial Research Organization (CSIRO), Ecosciences PrecinctDutton ParkQueenslandAustralia
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational LaboratoryGriffith Institute for Drug Discovery – Griffith UniversityBrisbaneQueenslandAustralia
- Menzies Health Institute, Griffith UniversitySouthportQueenslandAustralia
- Translational Research InstituteWoolloongabbaQueenslandAustralia
| |
Collapse
|
78
|
Królicki L, Kunikowska J, Bruchertseifer F, Kuliński R, Pawlak D, Koziara H, Rola R, Morgenstern A, Merlo A. Locoregional Treatment of Glioblastoma With Targeted α Therapy: [ 213 Bi]Bi-DOTA-Substance P Versus [ 225 Ac]Ac-DOTA-Substance P-Analysis of Influence Parameters. Clin Nucl Med 2023; 48:387-392. [PMID: 36854309 DOI: 10.1097/rlu.0000000000004608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
BACKGROUND Glioblastoma (GB) is the most malignant primary brain tumor. Therefore, introduction of new treatment options is critically important. The aim of this study was to assess local treatment with α emitters [ 213 Bi]Bi-DOTA-substance P (SP) and [ 225 Ac]Ac-DOTA-SP. METHODS Treatment was performed as salvage therapy in patients with recurrent primary and secondary GB. [ 213 Bi]Bi-DOTA-SP with injected activity 1.85 GBq per cycle was used in 20 primary (48.2 ± 11.8 years old) and in 9 secondary (38.8 ± 10.8 years old) GB patients and [ 225 Ac]Ac-DOTA-SP in 15 primary (45.1 ± 9.9 years old) and in 6 secondary (37.8 ± 6.4 years old) GB patients with a dose escalation scheme (10, 20, and 30 MBq). RESULTS Local treatment with [ 213 Bi]Bi-DOTA-SP and [ 225 Ac]Ac-DOTA-SP was well tolerated with only few adverse effects. There was no statistically significant difference between [ 213 Bi]Bi-DOTA-SP and [ 225 Ac]Ac-DOTA-SP groups in survival parameters. For primary GB, survival parameters of patients treated with [ 213 Bi]Bi-DOTA-SP and [ 225 Ac]Ac-DOTA-SP were as follows(in months): progression-free survival time, 2.7 versus 2.4; OS-d (overall survival from time of diagnosis to death from any cause), 23.6 versus 21.0; OS-t (overall survival from the start of treatment to death from any cause), 7.5 versus 5.0; and OS-r (overall survival from recurrence in primary tumors to death from any cause), 10.9 versus 12.0. Survival parameters of secondary GB patients treated with [ 213 Bi]Bi-DOTA-SP and [ 225 Ac]Ac-DOTA-SP were as follows (in months): progression-free survival time, 5.8 versus 2.4; OS-d, 52.3 versus 65.0; OS-t, 16.4 versus 16.0; and OS-c (overall survival from conversion into secondary GB multiforme to death from any cause), 18.4 versus 36.0. CONCLUSIONS The similarity results of 213 Bi or 225 Ac may suggest that the local treatment of brain tumors can be greatly simplified. The experience to date shows that local radioisotope treatment of brain tumors requires further dosimetry studies, taking into account the complexity of biological processes.
Collapse
Affiliation(s)
- Leszek Królicki
- From the Department of Nuclear Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Jolanta Kunikowska
- From the Department of Nuclear Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Radosław Kuliński
- From the Department of Nuclear Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Dariusz Pawlak
- Radioisotope Centre POLATOM, National Centre for Nuclear Research, Otwock
| | - Henryk Koziara
- Department of Neurosurgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw
| | - Rafał Rola
- Department of Neurology, Military Institute of Aviation Medicine, Warsaw, Poland
| | | | - Adrian Merlo
- Neurosurgical Center Berne and University of Basel, Switzerland
| |
Collapse
|
79
|
Kalita O, Kazda T, Reguli S, Jancalek R, Fadrus P, Slachta M, Pospisil P, Krska L, Vrbkova J, Hrabalek L, Smrcka M, Lipina R. Effects of Reoperation Timing on Survival among Recurrent Glioblastoma Patients: A Retrospective Multicentric Descriptive Study. Cancers (Basel) 2023; 15:cancers15092530. [PMID: 37173996 PMCID: PMC10177480 DOI: 10.3390/cancers15092530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Glioblastoma inevitably recurs, but no standard regimen has been established for treating this recurrent disease. Several reports claim that reoperative surgery can improve survival, but the effects of reoperation timing on survival have rarely been investigated. We, therefore, evaluated the relationship between reoperation timing and survival in recurrent GBM. A consecutive cohort of unselected patients (real-world data) from three neuro-oncology cancer centers was analyzed (a total of 109 patients). All patients underwent initial maximal safe resection followed by treatment according to the Stupp protocol. Those meeting the following criteria during progression were indicated for reoperation and were further analyzed in this study: (1) The tumor volume increased by >20-30% or a tumor was rediscovered after radiological disappearance; (2) The patient's clinical status was satisfactory (KS ≥ 70% and PS WHO ≤ gr. 2); (3) The tumor was localized without multifocality; (4) The minimum expected tumor volume reduction was above 80%. A univariate Cox regression analysis of postsurgical survival (PSS) revealed a statistically significant effect of reoperation on PSS from a threshold of 16 months after the first surgery. Cox regression models that stratified the Karnofsky score with age adjustment confirmed a statistically significant improvement in PSS for time-to-progression (TTP) thresholds of 22 and 24 months. The patient groups exhibiting the first recurrence at 22 and 24 months had better survival rates than those exhibiting earlier recurrences. For the 22-month group, the HR was 0.5 with a 95% CI of (0.27, 0.96) and a p-value of 0.036. For the 24-month group, the HR was 0.5 with a 95% CI of (0.25, 0.96) and a p-value of 0.039. Patients with the longest survival were also the best candidates for repeated surgery. Later recurrence of glioblastoma was associated with higher survival rates after reoperation.
Collapse
Affiliation(s)
- Ondrej Kalita
- Department of Neurosurgery, Faculty of Medicine and Dentistry, Palacky University in Olomouc, University Hospital Olomouc, Zdravotníků 248/7, 779 00 Olomouc, Czech Republic
- Department of Health Care Science, Faculty of Humanities, T. Bata University in Zlin, Stefanikova 5670, 760 01 Zlín, Czech Republic
| | - Tomas Kazda
- Department of Radiation Oncology, Faculty of Medicine, Masaryk University, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53 Brno, Czech Republic
| | - Stefan Reguli
- Department of Neurosurgery, Faculty of Medicine, University of Ostrava, University Hospital Ostrava, 17. Listopadu 1790/5, 708 52 Ostrava, Czech Republic
| | - Radim Jancalek
- Department of Neurosurgery, Faculty of Medicine, Masaryk University, St. Anne's University Hospital in Brno, Pekarska 664/53, 602 00 Brno, Czech Republic
| | - Pavel Fadrus
- Department of Neurosurgery, Faculty of Medicine, Masaryk University, University Hospital Brno, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Marek Slachta
- Department of Neurosurgery, Faculty of Medicine and Dentistry, Palacky University in Olomouc, University Hospital Olomouc, Zdravotníků 248/7, 779 00 Olomouc, Czech Republic
| | - Petr Pospisil
- Department of Radiation Oncology, Faculty of Medicine, Masaryk University, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656 53 Brno, Czech Republic
| | - Lukas Krska
- Department of Neurosurgery, Faculty of Medicine, University of Ostrava, University Hospital Ostrava, 17. Listopadu 1790/5, 708 52 Ostrava, Czech Republic
| | - Jana Vrbkova
- Institute of Molecular and Translate Medicine, Faculty of Medicine and Dentistry, Palacky University in Olomouc, Hnevotinska 133/5, 779 00 Olomouc, Czech Republic
| | - Lumir Hrabalek
- Department of Neurosurgery, Faculty of Medicine and Dentistry, Palacky University in Olomouc, University Hospital Olomouc, Zdravotníků 248/7, 779 00 Olomouc, Czech Republic
| | - Martin Smrcka
- Department of Neurosurgery, Faculty of Medicine, Masaryk University, University Hospital Brno, Jihlavská 20, 625 00 Brno, Czech Republic
| | - Radim Lipina
- Department of Neurosurgery, Faculty of Medicine, University of Ostrava, University Hospital Ostrava, 17. Listopadu 1790/5, 708 52 Ostrava, Czech Republic
| |
Collapse
|
80
|
Colardo M, Gargano D, Russo M, Petraroia M, Pensabene D, D'Alessandro G, Santoro A, Limatola C, Segatto M, Di Bartolomeo S. Bromodomain and Extraterminal Domain (BET) Protein Inhibition Hinders Glioblastoma Progression by Inducing Autophagy-Dependent Differentiation. Int J Mol Sci 2023; 24:ijms24087017. [PMID: 37108181 PMCID: PMC10138987 DOI: 10.3390/ijms24087017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive type of malignant primary brain tumor, and it is characterized by a high recurrence incidence and poor prognosis due to the presence of a highly heterogeneous mass of stem cells with self-renewal capacity and stemness maintenance ability. In recent years, the epigenetic landscape of GBM has been explored and many epigenetic alterations have been investigated. Among the investigated epigenetic abnormalities, the bromodomain and extra-terminal domain (BET) chromatin readers have been found to be significantly overexpressed in GBM. In this work, we investigated the effects of BET protein inhibition on GBM cell reprogramming. We found that the pan-BET pharmacological inhibitor JQ1 was able to promote a differentiation program in GBM cells, thus impairing cell proliferation and enhancing the toxicity of the drug Temozolomide (TMZ). Notably, the pro-differentiation capability of JQ1 was prevented in autophagy-defective models, suggesting that autophagy activation is necessary for BET protein activity in regulating glioma cell fate. Given the growing interest in epigenetic therapy, our results further support the possibility of introducing a BET-based approach in GBM clinical management.
Collapse
Affiliation(s)
- Mayra Colardo
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy
| | - Deborah Gargano
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy
| | - Miriam Russo
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy
| | - Michele Petraroia
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy
| | | | - Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, 00185 Rome, Italy
- Neuromed IRCCS, Via Atinense, 86077 Pozzilli, Italy
| | - Antonio Santoro
- Department of Human Neuroscience, Sapienza University of Rome, 00185 Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, 00185 Rome, Italy
- Neuromed IRCCS, Via Atinense, 86077 Pozzilli, Italy
| | - Marco Segatto
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy
| | | |
Collapse
|
81
|
Patel PD, Patel NV, Danish SF. The Evolution of Laser-Induced Thermal Therapy for the Treatment of Gliomas. Neurosurg Clin N Am 2023; 34:199-207. [PMID: 36906327 DOI: 10.1016/j.nec.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Laser-induced thermal therapy (LITT) has evolved over the past two decades to treat a number of intracranial pathologies. Although it initially emerged as a salvage treatment of surgically inoperable tumors or recurrent lesions that had exhausted more conventional treatments, it is now being used as a primary, first-line treatment in certain instances with outcomes comparable to traditional surgical resection. The authors discuss the evolution of LITT in the treatment of gliomas and future directions, which may further enhance the efficacy of this procedure.
Collapse
Affiliation(s)
- Purvee D Patel
- Department of Neurosurgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health - Jersey Shore University Medical Center, Nutley, NJ 07110, USA; Department of Neurosurgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health, Jersey Shore University Hospital, Jersey Shore University Medical Center, 19 Davis Avenue, Hope Tower 4th Floor, Neptune, NJ 07753, USA
| | - Nitesh V Patel
- Department of Neurosurgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health - Jersey Shore University Medical Center, Nutley, NJ 07110, USA; Department of Neurosurgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health, Jersey Shore University Hospital, Jersey Shore University Medical Center, 19 Davis Avenue, Hope Tower 4th Floor, Neptune, NJ 07753, USA
| | - Shabbar F Danish
- Department of Neurosurgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health - Jersey Shore University Medical Center, Nutley, NJ 07110, USA; Department of Neurosurgery, Hackensack Meridian School of Medicine, Hackensack Meridian Health, Jersey Shore University Hospital, Jersey Shore University Medical Center, 19 Davis Avenue, Hope Tower 4th Floor, Neptune, NJ 07753, USA.
| |
Collapse
|
82
|
Nam Y, Koo H, Yang Y, Shin S, Zhu Z, Kim D, Cho HJ, Mu Q, Choi SW, Sa JK, Seo YJ, Kim Y, Lee K, Oh JW, Kwon YJ, Park WY, Kong DS, Seol HJ, Lee JI, Park CK, Lee HW, Yoon Y, Wang J. Pharmacogenomic profiling reveals molecular features of chemotherapy resistance in IDH wild-type primary glioblastoma. Genome Med 2023; 15:16. [PMID: 36915208 PMCID: PMC10010007 DOI: 10.1186/s13073-023-01165-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Although temozolomide (TMZ) has been used as a standard adjuvant chemotherapeutic agent for primary glioblastoma (GBM), treating isocitrate dehydrogenase wild-type (IDH-wt) cases remains challenging due to intrinsic and acquired drug resistance. Therefore, elucidation of the molecular mechanisms of TMZ resistance is critical for its precision application. METHODS We stratified 69 primary IDH-wt GBM patients into TMZ-resistant (n = 29) and sensitive (n = 40) groups, using TMZ screening of the corresponding patient-derived glioma stem-like cells (GSCs). Genomic and transcriptomic features were then examined to identify TMZ-associated molecular alterations. Subsequently, we developed a machine learning (ML) model to predict TMZ response from combined signatures. Moreover, TMZ response in multisector samples (52 tumor sectors from 18 cases) was evaluated to validate findings and investigate the impact of intra-tumoral heterogeneity on TMZ efficacy. RESULTS In vitro TMZ sensitivity of patient-derived GSCs classified patients into groups with different survival outcomes (P = 1.12e-4 for progression-free survival (PFS) and 3.63e-4 for overall survival (OS)). Moreover, we found that elevated gene expression of EGR4, PAPPA, LRRC3, and ANXA3 was associated to intrinsic TMZ resistance. In addition, other features such as 5-aminolevulinic acid negative, mesenchymal/proneural expression subtypes, and hypermutation phenomena were prone to promote TMZ resistance. In contrast, concurrent copy-number-alteration in PTEN, EGFR, and CDKN2A/B was more frequent in TMZ-sensitive samples (Fisher's exact P = 0.0102), subsequently consolidated by multi-sector sequencing analyses. Integrating all features, we trained a ML tool to segregate TMZ-resistant and sensitive groups. Notably, our method segregated IDH-wt GBM patients from The Cancer Genome Atlas (TCGA) into two groups with divergent survival outcomes (P = 4.58e-4 for PFS and 3.66e-4 for OS). Furthermore, we showed a highly heterogeneous TMZ-response pattern within each GBM patient using in vitro TMZ screening and genomic characterization of multisector GSCs. Lastly, the prediction model that evaluates the TMZ efficacy for primary IDH-wt GBMs was developed into a webserver for public usage ( http://www.wang-lab-hkust.com:3838/TMZEP ). CONCLUSIONS We identified molecular characteristics associated to TMZ sensitivity, and illustrate the potential clinical value of a ML model trained from pharmacogenomic profiling of patient-derived GSC against IDH-wt GBMs.
Collapse
Affiliation(s)
- Yoonhee Nam
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Harim Koo
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, South Korea
- Department of Clinical Research, Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Yingxi Yang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Sang Shin
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| | - Zhihan Zhu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Donggeon Kim
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| | - Hee Jin Cho
- Department of Biomedical Convergence Science and Technology, School of Convergence, Kyungpook National University, Daegu, South Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, South Korea
| | - Quanhua Mu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Seung Won Choi
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University, New York, NY, USA
| | - Jason K Sa
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Yun Jee Seo
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| | - Yejin Kim
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| | - Kyoungmin Lee
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| | - Jeong-Woo Oh
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| | - Yong-Jun Kwon
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| | - Woong-Yang Park
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Doo-Sik Kong
- Department of Neurosurgery, Samsung Medical Center and Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ho Jun Seol
- Department of Neurosurgery, Samsung Medical Center and Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jung-Il Lee
- Department of Neurosurgery, Samsung Medical Center and Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Chul-Kee Park
- Department of Neurosurgery, College of Medicine, Seoul National University and Seoul National University Hospital, Seoul, South Korea
| | - Hye Won Lee
- Department of Urology, Center for Urologic Cancer, National Cancer Center, Goyang, South Korea.
- Department of Urology, Samsung Medical Center, Seoul, South Korea.
| | - Yeup Yoon
- Institute for Refractory Cancer Research, Research Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea.
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea.
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Seoul, South Korea.
| | - Jiguang Wang
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
- Hong Kong Center for Neurodegenerative Diseases, InnoHK, Hong Kong SAR, China.
- HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen, China.
| |
Collapse
|
83
|
Salerno S, Barresi E, Baglini E, Poggetti V, Da Settimo F, Taliani S. Target-Based Anticancer Indole Derivatives for the Development of Anti-Glioblastoma Agents. Molecules 2023; 28:molecules28062587. [PMID: 36985576 PMCID: PMC10056347 DOI: 10.3390/molecules28062587] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive and frequent primary brain tumor, with a poor prognosis and the highest mortality rate. Currently, GBM therapy consists of surgical resection of the tumor, radiotherapy, and adjuvant chemotherapy with temozolomide. Consistently, there are poor treatment options and only modest anticancer efficacy is achieved; therefore, there is still a need for the development of new effective therapies for GBM. Indole is considered one of the most privileged scaffolds in heterocyclic chemistry, so it may serve as an effective probe for the development of new drug candidates against challenging diseases, including GBM. This review analyzes the therapeutic benefit and clinical development of novel indole-based derivatives investigated as promising anti-GBM agents. The existing indole-based compounds which are in the pre-clinical and clinical stages of development against GBM are reported, with particular reference to the most recent advances between 2013 and 2022. The main mechanisms of action underlying their anti-GBM efficacy, such as protein kinase, tubulin and p53 pathway inhibition, are also discussed. The final goal is to pave the way for medicinal chemists in the future design and development of novel effective indole-based anti-GBM agents.
Collapse
|
84
|
Martino T. Novel baseline biomarkers should predict recurrence of glioblastoma. JOURNAL OF CLINICAL ULTRASOUND : JCU 2023; 51:571-573. [PMID: 36893042 DOI: 10.1002/jcu.23401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 06/18/2023]
Affiliation(s)
- Tommaso Martino
- Neuroscience Department (Head of Department: Dr. Ciro Mundi), Policlinico Riuniti of Foggia, S.C. Ospedaliera di Neurologia, Foggia, Italy
| |
Collapse
|
85
|
Prajapati HP, Ansari A. Updates in the Management of Recurrent Glioblastoma Multiforme. J Neurol Surg A Cent Eur Neurosurg 2023; 84:174-187. [PMID: 35772723 DOI: 10.1055/s-0042-1749351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND Glioblastoma is the most aggressive and diffusely infiltrative primary brain tumor. Recurrence is almost universal even after all primary standard treatments. This article aims to review the literature and update the standard treatment strategies for patients with recurrent glioblastoma. METHODS A systematic search was performed with the phrase "recurrent glioblastoma and management" as a search term in PubMed central, Medline, and Embase databases to identify all the articles published on the subject till December 2020. The review included peer-reviewed original articles, clinical trials, review articles, and keywords in title and abstract. RESULTS Out of 513 articles searched, 73 were included in this review after screening for eligibility. On analyzing the data, most of the studies report a median overall survival (OS) of 5.9 to 11.4 months after re-surgery and 4.7 to 7.6 months without re-surgery. Re-irradiation with stereotactic radiosurgery (SRS) and fractionated stereotactic radiotherapy (FSRT) result in a median OS of 10.2 months (range: 7.0-12 months) and 9.8 months (ranged: 7.5-11.0 months), respectively. Radiation necrosis was found in 16.6% (range: 0-24.4%) after SRS. Chemotherapeutic agents like nitrosourea (carmustine), bevacizumab, and temozolomide (TMZ) rechallenge result in a median OS in the range of 5.1 to 7.5, 6.5 to 9.2, and 5.1-13.0 months and six months progression free survival (PFS-6) in the range of 13 to 17.5%, 25 to 42.6%, and 23 to 58.3%, respectively. Use of epithelial growth factor receptor (EGFR) inhibitors results in a median OS in the range of 2.0 to 3.0 months and PFS-6 in 13%. CONCLUSION Although recurrent glioblastoma remains a fatal disease with universal mortality, the literature suggests that a subset of patients may benefit from maximal treatment efforts.
Collapse
Affiliation(s)
- Hanuman Prasad Prajapati
- Department of Neurosurgery, Uttar Pradesh University of Medical Sciences, Etawah, Uttar Pradesh, India
| | - Ahmad Ansari
- Department of Neurosurgery, Uttar Pradesh University of Medical Sciences, Safai, Uttar Pradesh, India
| |
Collapse
|
86
|
Wu H, Guo C, Wang C, Xu J, Zheng S, Duan J, Li Y, Bai H, Xu Q, Ning F, Wang F, Yang Q. Single-cell RNA sequencing reveals tumor heterogeneity, microenvironment, and drug-resistance mechanisms of recurrent glioblastoma. Cancer Sci 2023. [PMID: 36853018 DOI: 10.1111/cas.15773] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/02/2023] [Accepted: 02/20/2023] [Indexed: 03/01/2023] Open
Abstract
Glioblastomas are highly heterogeneous brain tumors. Despite the availability of standard treatment for glioblastoma multiforme (GBM), i.e., Stupp protocol, which involves surgical resection followed by radiotherapy and chemotherapy, glioblastoma remains refractory to treatment and recurrence is inevitable. Moreover, the biology of recurrent glioblastoma remains unclear. Increasing evidence has shown that intratumoral heterogeneity and the tumor microenvironment contribute to therapeutic resistance. However, the interaction between intracellular heterogeneity and drug resistance in recurrent GBMs remains controversial. The aim of this study was to map the transcriptome landscape of cancer cells and the tumor heterogeneity and tumor microenvironment in recurrent and drug-resistant GBMs at a single-cell resolution and further explore the mechanism of drug resistance of GBMs. We analyzed six tumor tissue samples from three patients with primary GBM and three patients with recurrent GBM in which recurrence and drug resistance developed after treatment with the standard Stupp protocol using single-cell RNA sequencing. Using unbiased clustering, nine major cell clusters were identified. Upregulation of the expression of stemness-related and cell-cycle-related genes was observed in recurrent GBM cells. Compared with the initial GBM tissues, recurrent GBM tissues showed a decreased proportion of microglia, consistent with previous reports. Finally, vascular endothelial growth factor A expression and the blood-brain barrier permeability were high, and the O6 -methylguanine DNA methyltransferase-related signaling pathway was activated in recurrent GBM. Our results delineate the single-cell map of recurrent glioblastoma, tumor heterogeneity, tumor microenvironment, and drug-resistance mechanisms, providing new insights into treatment strategies for recurrent glioblastomas.
Collapse
Affiliation(s)
- Haibin Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chengcheng Guo
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chaoye Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Biometric Information, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiang Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Suyue Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Duan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiyun Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongming Bai
- Department of Neurosurgery, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
| | - Qiuyan Xu
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fangling Ning
- Department of Medical Oncology, Binzhou Medical University Hospital, Binzhou, China
| | - Feng Wang
- Department of Medical Oncology, Binzhou Medical University Hospital, Binzhou, China
| | - Qunying Yang
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
87
|
Bian Y, Wang Y, Chen X, Zhang Y, Xiong S, Su D. Image‐guided diagnosis and treatment of glioblastoma. VIEW 2023. [DOI: 10.1002/viw.20220069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Yongning Bian
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Yaling Wang
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Xueqian Chen
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Yong Zhang
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Shaoqing Xiong
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| | - Dongdong Su
- Center of Excellence for Environmental Safety and Biological Effects Beijing Key Laboratory for Green Catalysis and Separation Department of Chemistry Beijing University of Technology Beijing P. R. China
| |
Collapse
|
88
|
Perwein T, Giese B, Nussbaumer G, von Bueren AO, van Buiren M, Benesch M, Kramm CM. How I treat recurrent pediatric high-grade glioma (pHGG): a Europe-wide survey study. J Neurooncol 2023; 161:525-538. [PMID: 36720762 PMCID: PMC9992031 DOI: 10.1007/s11060-023-04241-6] [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: 11/30/2022] [Accepted: 01/05/2023] [Indexed: 02/02/2023]
Abstract
PURPOSE As there is no standard of care treatment for recurrent/progressing pediatric high-grade gliomas (pHGG), we aimed to gain an overview of different treatment strategies. METHODS In a web-based questionnaire, members of the SIOPE-BTG and the GPOH were surveyed on therapeutic options in four case scenarios (children/adolescents with recurrent/progressing HGG). RESULTS 139 clinicians with experience in pediatric neuro-oncology from 22 European countries participated in the survey. Most respondents preferred further oncological treatment in three out of four cases and chose palliative care in one case with marked symptoms. Depending on the case, 8-92% would initiate a re-resection (preferably hemispheric pHGG), combined with molecular diagnostics. Throughout all case scenarios, 55-77% recommended (re-)irradiation, preferably local radiotherapy > 20 Gy. Most respondents would participate in clinical trials and use targeted therapy (79-99%), depending on molecular genetic findings (BRAF alterations: BRAF/MEK inhibitor, 64-88%; EGFR overexpression: anti-EGFR treatment, 46%; CDKN2A deletion: CDK inhibitor, 18%; SMARCB1 deletion: EZH2 inhibitor, 12%). 31-72% would administer chemotherapy (CCNU, 17%; PCV, 8%; temozolomide, 19%; oral etoposide/trofosfamide, 8%), and 20-69% proposed immunotherapy (checkpoint inhibitors, 30%; tumor vaccines, 16%). Depending on the individual case, respondents would also include bevacizumab (6-18%), HDAC inhibitors (4-15%), tumor-treating fields (1-26%), and intraventricular chemotherapy (4-24%). CONCLUSION In each case, experts would combine conventional multimodal treatment concepts, including re-irradiation, with targeted therapy based on molecular genetic findings. International cooperative trials combining a (chemo-)therapy backbone with targeted therapy approaches for defined subgroups may help to gain valid clinical data and improve treatment in pediatric patients with recurrent/progressing HGG.
Collapse
Affiliation(s)
- Thomas Perwein
- Division of Pediatric Hemato-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Auenbruggerplatz 34/2, 8036, Graz, Austria.
| | - Barbara Giese
- Division of Pediatric Hemato-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Auenbruggerplatz 34/2, 8036, Graz, Austria
| | - Gunther Nussbaumer
- Division of Pediatric Hemato-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Auenbruggerplatz 34/2, 8036, Graz, Austria
| | - André O von Bueren
- Department of Pediatrics, Obstetrics and Gynecology, Division of Pediatric Hematology and Oncology, University Hospital of Geneva, Geneva, Switzerland
- Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of Medicine, Department of Pediatrics, Gynecology and Obstetrics, University of Geneva, Geneva, Switzerland
| | - Miriam van Buiren
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, Medical Center, University of Freiburg, Freiburg, Germany
| | - Martin Benesch
- Division of Pediatric Hemato-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, Auenbruggerplatz 34/2, 8036, Graz, Austria
| | - Christof Maria Kramm
- Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
89
|
Mejía-Rodríguez R, Romero-Trejo D, González RO, Segovia J. Combined treatments with AZD5363, AZD8542, curcumin or resveratrol induce death of human glioblastoma cells by suppressing the PI3K/AKT and SHH signaling pathways. Biochem Biophys Rep 2023; 33:101430. [PMID: 36714540 PMCID: PMC9876780 DOI: 10.1016/j.bbrep.2023.101430] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/02/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma (GBM) is a very aggressive tumor that presents vascularization, necrosis and is resistant to chemotherapy and radiotherapy. Current treatments are not effective eradicating GBM, thus, there is an urgent need to develop novel therapeutic strategies against GBM. AZD5363, AZD8542, curcumin and resveratrol, are widely studied for the treatment of cancer and in the present study we explored the effects of the administration of combined treatments with AZD5363, AZD8542, curcumin or resveratrol on human GBM cells. We found that the combined treatments with AZD5363+AZD8542+Curcumin and AZD8542+Curcumin+Resveratrol inhibit the PI3K/AKT and SHH survival pathways by decreasing the activity of AKT, the reduction of the expression of SMO, pP70S6k, pS6k, GLI1, p21 and p27, and the activation of caspase-3 as a marker of apoptosis. These results provide evidence that the combined treatments AZD5363+AZD8542+Curcumin and AZD8542+Curcumin+Resveratrol have the potential to be an interesting option against GBM.
Collapse
Affiliation(s)
- Rosalinda Mejía-Rodríguez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico
| | - Daniel Romero-Trejo
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico
| | - Rosa O. González
- Departamento de Matemáticas, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), Mexico
| | - José Segovia
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico,Corresponding author. Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN # 2508, 07300, Mexico.
| |
Collapse
|
90
|
Kebir S, Ullrich V, Berger P, Dobersalske C, Langer S, Rauschenbach L, Trageser D, Till A, Lorbeer FK, Wieland A, Wilhelm-Buchstab T, Ahmad A, Fröhlich H, Cima I, Prasad S, Matschke J, Jendrossek V, Remke M, Grüner BM, Roesch A, Siveke JT, Herold-Mende C, Blau T, Keyvani K, van Landeghem FK, Pietsch T, Felsberg J, Reifenberger G, Weller M, Sure U, Brüstle O, Simon M, Glas M, Scheffler B. A Sequential Targeting Strategy Interrupts AKT-Driven Subclone-Mediated Progression in Glioblastoma. Clin Cancer Res 2023; 29:488-500. [PMID: 36239995 PMCID: PMC9843437 DOI: 10.1158/1078-0432.ccr-22-0611] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 09/10/2022] [Accepted: 10/07/2022] [Indexed: 01/21/2023]
Abstract
PURPOSE Therapy resistance and fatal disease progression in glioblastoma are thought to result from the dynamics of intra-tumor heterogeneity. This study aimed at identifying and molecularly targeting tumor cells that can survive, adapt, and subclonally expand under primary therapy. EXPERIMENTAL DESIGN To identify candidate markers and to experimentally access dynamics of subclonal progression in glioblastoma, we established a discovery cohort of paired vital cell samples obtained before and after primary therapy. We further used two independent validation cohorts of paired clinical tissues to test our findings. Follow-up preclinical treatment strategies were evaluated in patient-derived xenografts. RESULTS We describe, in clinical samples, an archetype of rare ALDH1A1+ tumor cells that enrich and acquire AKT-mediated drug resistance in response to standard-of-care temozolomide (TMZ). Importantly, we observe that drug resistance of ALDH1A1+ cells is not intrinsic, but rather an adaptive mechanism emerging exclusively after TMZ treatment. In patient cells and xenograft models of disease, we recapitulate the enrichment of ALDH1A1+ cells under the influence of TMZ. We demonstrate that their subclonal progression is AKT-driven and can be interfered with by well-timed sequential rather than simultaneous antitumor combination strategy. CONCLUSIONS Drug-resistant ALDH1A1+/pAKT+ subclones accumulate in patient tissues upon adaptation to TMZ therapy. These subclones may therefore represent a dynamic target in glioblastoma. Our study proposes the combination of TMZ and AKT inhibitors in a sequential treatment schedule as a rationale for future clinical investigation.
Collapse
Affiliation(s)
- Sied Kebir
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, Essen, Germany
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Vivien Ullrich
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Pia Berger
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Celia Dobersalske
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Langer
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Laurèl Rauschenbach
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, Essen, Germany
| | - Daniel Trageser
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany
| | - Andreas Till
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Franziska K. Lorbeer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | - Anja Wieland
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
| | | | - Ashar Ahmad
- Bonn-Aachen International Center for IT (B-IT), University of Bonn, Bonn, Germany
| | - Holger Fröhlich
- Bonn-Aachen International Center for IT (B-IT), University of Bonn, Bonn, Germany
- Department of Bioinformatics, Fraunhofer SCAI, Schloss Birlinghoven, Sankt Augustin, Germany
| | - Igor Cima
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Shruthi Prasad
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Marc Remke
- German Cancer Consortium (DKTK)
- Pediatric Neuro-Oncogenomics, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Barbara M. Grüner
- German Cancer Consortium (DKTK)
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Alexander Roesch
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Department of Dermatology, University Hospital Essen, Essen, Germany
| | - Jens T. Siveke
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Christel Herold-Mende
- Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Tobias Blau
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University of Duisburg-Essen, Essen, Germany
| | | | - Torsten Pietsch
- Institute of Neuropathology, University of Bonn, Bonn, Germany
| | - Jörg Felsberg
- Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Guido Reifenberger
- German Cancer Consortium (DKTK)
- Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Ulrich Sure
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- Department of Neurosurgery and Spine Surgery, University Hospital Essen, Essen, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany
| | - Matthias Simon
- Department of Neurosurgery, University of Bonn Medical Center, Bonn, Germany
- Department of Neurosurgery, Bethel Clinic, University of Bielefeld Medical Center, OWL, Bielefeld, Germany
| | - Martin Glas
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, Essen, Germany
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
| | - Björn Scheffler
- DKFZ-Division Translational Neurooncology at the WTZ, DKTK Partner Site, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK)
- West German Cancer Center (WTZ), University Hospital Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Center of Medical Biotechnology (ZMB), University Duisburg-Essen, Essen, Germany
- Corresponding Author: Björn Scheffler, Professor for Translational Oncology, DKFZ-Division of Translational Neurooncology at the West German Cancer Center (WTZ), DKTK Partner Site, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, WTZ-F, UG 01.041, Essen D-45147, Germany. Phone: 49 (0)201-723-8130; Fax: 49 (0)201-723-6752; E-mail:
| |
Collapse
|
91
|
She L, Gong X, Su L, Liu C. Effectiveness and safety of tumor-treating fields therapy for glioblastoma: A single-center study in a Chinese cohort. Front Neurol 2023; 13:1042888. [PMID: 36698900 PMCID: PMC9869119 DOI: 10.3389/fneur.2022.1042888] [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: 09/13/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Objective Tumor-treating fields (TTFields) are a new therapeutic modality for patients with glioblastoma (GBM). However, studies on survival outcomes of TTFields are rarely reported in China. This study aimed to examine the clinical efficacy and safety of TTFields therapy for GBM in China. Methods A total of 93 patients with newly diagnosed GBM (ndGBM) and recurrent GBM (rGBM) were included in our study retrospectively. They were divided into two groups based on whether they used TTFields. Progression-free survival (PFS), overall survival (OS), and toxicities were assessed. Results Among the patients with ndGBM, there were 13 cases with TTFields and 39 cases with no TTFields. The median PFS was 15.3 [95% confidence interval (CI): 6.5-24.1] months and 10.6 (95% CI: 5.4-15.8) months in the two groups, respectively, with P = 0.041. The median OS was 24.8 (95% CI: 6.8-42.8) months and 18.6 (95% CI: 11.4-25.8) months, respectively, with P = 0.368. Patients with subtotal resection (STR) who used TTFields had a better PFS than those who did not (P = 0.003). Among the patients with rGBM, there were 13 cases with TTFields and 28 cases with no TTFields. The median PFS in the two groups was 8.4 (95% CI: 1.7-15.2) months and 8.0 (95% CI: 5.8-10.2) months in the two groups, respectively, with P = 0.265. The median OS was 10.6 (95% CI: 4.8-16.4) months and 13.3 (95% CI: 11.0-15.6) months, respectively, with P = 0.655. A total of 21 patients (21/26, 80.8%) with TTFields developed dermatological adverse events (dAEs). All the dAEs could be resolved or controlled. Conclusion TTFields therapy is a safe and effective treatment for ndGBM, especially in patients with STR. However, it may not improve survival in patients with rGBM.
Collapse
Affiliation(s)
- Lei She
- Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China,Hunan Key Laboratory of Pharmacogenetics, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuan Gong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Su
- Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chao Liu
- Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China,*Correspondence: Chao Liu ✉
| |
Collapse
|
92
|
Effectiveness of Flattening-Filter-Free versus Flattened Beams in V79 and Glioblastoma Patient-Derived Stem-like Cells. Int J Mol Sci 2023; 24:ijms24021107. [PMID: 36674623 PMCID: PMC9861147 DOI: 10.3390/ijms24021107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Literature data on the administration of conventional high-dose beams with (FF) or without flattening filters (FFF) show conflicting results on biological effects at the cellular level. To contribute to this field, we irradiated V79 Chinese hamster lung fibroblasts and two patient-derived glioblastoma stem-like cell lines (GSCs-named #1 and #83) using a clinical 10 MV accelerator with FF (at 4 Gy/min) and FFF (at two dose rates 4 and 24 Gy/min). Cell killing and DNA damage induction, determined using the γ-H2AX assay, and gene expression were studied. No significant differences in the early survival of V79 cells were observed as a function of dose rates and FF or FFF beams, while a trend of reduction in late survival was observed at the highest dose rate with the FFF beam. GSCs showed similar survival levels as a function of dose rates, both delivered in the FFF regimen. The amount of DNA damage measured for both dose rates after 2 h was much higher in line #1 than in line #83, with statistically significant differences between the two dose rates only in line #83. The gene expression analysis of the two GSC lines indicates gene signatures mimicking the prognosis of glioblastoma (GBM) patients derived from a public database. Overall, the results support the current use of FFF and highlight the possibility of identifying patients with candidate gene signatures that could benefit from irradiation with FFF beams at a high dose rate.
Collapse
|
93
|
Yang W, Wang S, Zhang X, Sun H, Zhang M, Chen H, Cui J, Li J, Peng F, Zhu M, Yu B, Li Y, Yang L, Min W, Xue M, Pan L, Zhu H, Wu B, Gu Y. New natural compound inhibitors of PDGFRA (platelet-derived growth factor receptor α) based on computational study for high-grade glioma therapy. Front Neurosci 2023; 16:1060012. [PMID: 36685223 PMCID: PMC9845622 DOI: 10.3389/fnins.2022.1060012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 12/01/2022] [Indexed: 01/06/2023] Open
Abstract
Background High-grade glioma (HGG) is a malignant brain tumor that is common and aggressive in children and adults. In the current medical paradigm, surgery and radiotherapy are the standard treatments for HGG patients. Despite this, the overall prognosis is still very bleak. Studies have shown that platelet-derived growth factor receptor α (PDGFRA) is an essential target to treat tumors and inhibiting the activity of PDGFRA can improve the prognosis of HGG. Thus, PDGFRA inhibitors are critical to developing drugs and cancer treatment. Objective The purpose of this study was to screen lead compounds and candidate drugs with potential inhibitors against platelet-derived growth factor receptor α (PDGFRA) from the drug library (ZINC database) in order to improve the prognosis of patients with high-grade glioma (HGG). Materials and methods In our study, we selected Imatinib as the reference drug. A series of computer-aided technologies, such as Discovery Studio 2019 and Schrodinger, were used to screen and assess potential inhibitors of PDGFRA. The first step was to calculate the LibDock scores and then analyze the pharmacological and toxicological properties. Following this, we docked the small molecules selected in the previous steps with PDGFRA to study their docking mechanism and affinity. In addition, molecular dynamics simulation was used to determine whether the ligand-PDGFRA complex was stable in nature. Results Two novel natural compounds 1 and 2 (ZINC000008829785 and ZINC000013377891) from the ZINC database were found binding to PDGFRA with more favorable interaction energy. Also, they were predicted with less Ames mutagenicity, rodent carcinogenicity, non-developmental toxic potential, and tolerant with cytochrome P450 2D6 (CYP2D6). The dynamic simulation analysis demonstrated that ZINC000008829785-PDGFRA and ZINC000013377891-PDGFRA dimer complex had more favorable potential energy compared with Imatinib, and they can exist in natural environments stably. Conclusion ZINC000008829785 and ZINC000013377891 might provide a solid foundation for drugs that inhibit PDGFRA in HGG. In addition to being safe drug candidates, these compounds had important implications for improving drugs targeting PDGFRA.
Collapse
Affiliation(s)
- Wenzhuo Yang
- Department of Neurosurgery, Zibo Central Hospital, Zibo, China,Department of Neurosurgery, Cancer Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shengnan Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xiangmao Zhang
- Department of Neurosurgery, Zibo Central Hospital, Zibo, China
| | - Hu Sun
- Department of Neurosurgery, Zibo Central Hospital, Zibo, China
| | - Menghan Zhang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Xinxiang Medical College, Xinxiang, China
| | - Hongyu Chen
- Department of Neurosurgery, Cancer Hospital of Sun Yat-sen University, Guangzhou, China
| | - Junxiang Cui
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Jinyang Li
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Fei Peng
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Mingqin Zhu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Bingcheng Yu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yifan Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Liu Yang
- Department of Neurosurgical Oncology, The First Hospital of Jilin University, Changchun, China
| | - Wanwan Min
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Mengru Xue
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Lin Pan
- School of Clinical Medicine, Jilin University, Changchun, China
| | - Hao Zhu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Bo Wu
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, China
| | - Yinghao Gu
- Department of Neurosurgery, Zibo Central Hospital, Zibo, China,*Correspondence: Yinghao Gu,
| |
Collapse
|
94
|
DGKB mediates radioresistance by regulating DGAT1-dependent lipotoxicity in glioblastoma. Cell Rep Med 2023; 4:100880. [PMID: 36603576 PMCID: PMC9873821 DOI: 10.1016/j.xcrm.2022.100880] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/08/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023]
Abstract
Glioblastoma (GBM) currently has a dismal prognosis. GBM cells that survive radiotherapy contribute to tumor progression and recurrence with metabolic advantages. Here, we show that diacylglycerol kinase B (DGKB), a regulator of the intracellular concentration of diacylglycerol (DAG), is significantly downregulated in radioresistant GBM cells. The downregulation of DGKB increases DAG accumulation and decreases fatty acid oxidation, contributing to radioresistance by reducing mitochondrial lipotoxicity. Diacylglycerol acyltransferase 1 (DGAT1), which catalyzes the formation of triglycerides from DAG, is increased after ionizing radiation. Genetic inhibition of DGAT1 using short hairpin RNA (shRNA) or microRNA-3918 (miR-3918) mimic suppresses radioresistance. We discover that cladribine, a clinical drug, activates DGKB, inhibits DGAT1, and sensitizes GBM cells to radiotherapy in vitro and in vivo. Together, our study demonstrates that DGKB downregulation and DGAT1 upregulation confer radioresistance by reducing mitochondrial lipotoxicity and suggests DGKB and DGAT1 as therapeutic targets to overcome GBM radioresistance.
Collapse
|
95
|
Takacs GP, Kreiger CJ, Luo D, Tian G, Garcia JS, Deleyrolle LP, Mitchell DA, Harrison JK. Glioma-derived CCL2 and CCL7 mediate migration of immune suppressive CCR2 +/CX3CR1 + M-MDSCs into the tumor microenvironment in a redundant manner. Front Immunol 2023; 13:993444. [PMID: 36685592 PMCID: PMC9854274 DOI: 10.3389/fimmu.2022.993444] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common and malignant primary brain tumor, resulting in poor survival despite aggressive therapies. GBM is characterized in part by a highly heterogeneous and immunosuppressive tumor microenvironment (TME) made up predominantly of infiltrating peripheral immune cells. One significant immune cell type that contributes to glioma immune evasion is a population of immunosuppressive, hematopoietic cells, termed myeloid-derived suppressor cells (MDSCs). Previous studies suggest that a potent subset of myeloid cells, expressing monocytic (M)-MDSC markers, distinguished by dual expression of chemokine receptors CCR2 and CX3CR1, utilize CCR2 to infiltrate into the TME. This study evaluated the T cell suppressive function and migratory properties of CCR2+/CX3CR1+ MDSCs. Bone marrow-derived CCR2+/CX3CR1+ cells adopt an immune suppressive cell phenotype when cultured with glioma-derived factors. Recombinant and glioma-derived CCL2 and CCL7 induce the migration of CCR2+/CX3CR1+ MDSCs with similar efficacy. KR158B-CCL2 and -CCL7 knockdown murine gliomas contain equivalent percentages of CCR2+/CX3CR1+ MDSCs compared to KR158B gliomas. Combined neutralization of CCL2 and CCL7 completely blocks CCR2-expressing cell migration to KR158B cell conditioned media. CCR2+/CX3CR1+ cells are also reduced within KR158B gliomas upon combination targeting of CCL2 and CCL7. High levels of CCL2 and CCL7 are also associated with negative prognostic outcomes in GBM patients. These data provide a more comprehensive understanding of the function of CCR2+/CX3CR1+ MDSCs and the role of CCL2 and CCL7 in the recruitment of these immune suppressive cells and further support the significance of targeting this chemokine axis in GBM.
Collapse
Affiliation(s)
- Gregory P. Takacs
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Christian J. Kreiger
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Defang Luo
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Guimei Tian
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Julia S. Garcia
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Loic P. Deleyrolle
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Duane A. Mitchell
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jeffrey K. Harrison
- Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
96
|
Mantica M, Drappatz J. Immunotherapy associated central nervous system complications in primary brain tumors. Front Oncol 2023; 13:1124198. [PMID: 36874119 PMCID: PMC9981156 DOI: 10.3389/fonc.2023.1124198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/23/2023] [Indexed: 02/18/2023] Open
Abstract
Advances clarifying the genetics and function of the immune system within the central nervous system (CNS) and brain tumor microenvironment have led to increasing momentum and number of clinical trials using immunotherapy for primary brain tumors. While neurological complications of immunotherapy in extra-cranial malignancies is well described, the CNS toxicities of immunotherapy in patients with primary brain tumors with their own unique physiology and challenges are burgeoning. This review highlights the emerging and unique CNS complications associated with immunotherapy including checkpoint inhibitors, oncolytic viruses, adoptive cell transfer/chimeric antigen receptor (CAR) T cell and vaccines for primary brain tumors, as well as reviews modalities that have been currently employed or are undergoing investigation for treatment of such toxicities.
Collapse
Affiliation(s)
- Megan Mantica
- Department of Neurology, University of Pittsburgh, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Jan Drappatz
- Department of Neurology, University of Pittsburgh, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| |
Collapse
|
97
|
Vargas López AJ, Fernández Carballal C, Valera Melé M, Rodríguez-Boto G. Survival analysis in high-grade glioma: The role of salvage surgery. Neurologia 2023; 38:21-28. [PMID: 36464224 DOI: 10.1016/j.nrleng.2020.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 04/01/2020] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVES This study addresses the survival of consecutive patients with high-grade gliomas (HGG) treated at the same institution over a period of 10 years. We analyse the importance of associated factors and the role of salvage surgery at the time of progression. METHODS We retrospectively analysed a series of patients with World Health Organization (WHO) grade III/IV gliomas treated between 2008 and 2017 at Hospital Gregorio Marañón (Madrid, Spain). Clinical, radiological, and anatomical pathology data were obtained from patient clinical histories. RESULTS Follow-up was completed in 233 patients with HGG. Mean age was 62.2 years. The median survival time was 15.4 months. Of 133 patients (59.6%) who had undergone surgery at the time of diagnosis, 43 (32.3%) underwent salvage surgery at the time of progression. This subgroup presented longer overall survival and survival after progression. Higher Karnofsky Performance Status score at diagnosis, a greater extent of surgical resection, and initial diagnosis of WHO grade III glioma were also associated with longer survival. CONCLUSIONS About one-third of patients with HGG may be eligible for salvage surgery at the time of progression. Salvage surgery in this subgroup of patients was significantly associated with longer survival.
Collapse
Affiliation(s)
- A J Vargas López
- Servicio de Neurocirugía, Hospital Universitario Torrecárdenas, Almería, Spain; Programa de Doctorado en Medicina y Cirugía, Universidad Autónoma de Madrid, Madrid, Spain.
| | - C Fernández Carballal
- Servicio de Neurocirugía, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - M Valera Melé
- Servicio de Neurocirugía, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - G Rodríguez-Boto
- Programa de Doctorado en Medicina y Cirugía, Universidad Autónoma de Madrid, Madrid, Spain; Servicio de Neurocirugía, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| |
Collapse
|
98
|
Acharekar A, Bachal K, Shirke P, Thorat R, Banerjee A, Gardi N, Majumder A, Dutt S. Substrate stiffness regulates the recurrent glioblastoma cell morphology and aggressiveness. Matrix Biol 2023; 115:107-127. [PMID: 36563706 DOI: 10.1016/j.matbio.2022.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 12/14/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022]
Abstract
Recurrent glioblastoma is highly aggressive with currently no specific treatment regime. Therefore, to identify novel therapeutic targets for recurrent GBM, we used a cellular model developed in our lab from commercially available cell line U87MG and patient-derived cultures that allows the comparison between radiation naïve (Parent) and recurrent GBM cells generated after parent cells are exposed to lethal dose of radiation. Total RNA-seq of parent and recurrent population revealed significant upregulation of cell-ECM interactions pathway in the recurrent population. These results led us to hypothesize that the physical microenvironment contributes to the aggressiveness of recurrent GBM. To verify this, we cultured parent and recurrent GBM cells on collagen-coated polyacrylamide gels mimicking the stiffness of normal brain (Young's modulus E = 0.5kPa) or tumorigenic brain (E = 10kPa) and tissue culture plastic dishes (E ∼ 1 GPa). We found that compared to parent cells, recurrent cells showed higher proliferation, invasion, migration, and resistance to EGFR inhibitor. Using orthotopic GBM mouse model and resection model, we demonstrate that recurrent cells cultured on 0.5kPa had higher in vivo tumorigenicity and recurrent disease progression than parent cells, whereas these differences were insignificant when parent and recurrent cells were cultured on plastic substrates. Furthermore, recurrent cells on 0.5kPa showed high expression of ECM proteins like Collagen, MMP2 and MMP9. These proteins were also significantly upregulated in recurrent patient biopsies. Additionally, the brain of mice injected with recurrent cells grown on 0.5kPa showed higher Young's moduli suggesting the ability of these cells to make the surrounding ECM stiffer. Total RNA-seq of parent and recurrent cells grown on plastic and 0.5kpa identified PLEKHA7 significantly upregulated specifically in recurrent cells grown on 0.5 kPa substrate. PLEKHA7 was also found to be high in recurrent GBM patient biopsies. Accordingly, PLEKHA7 knockdown reduced invasion and survival of recurrent GBM cells. Together, these data provide an in vitro model system that captures the observed in vivo and clinical behavior of recurrent GBM by mimicking mechanical microenvironment and identifies PLEKHA7 as a novel potential target for recurrent GBM.
Collapse
Affiliation(s)
- Anagha Acharekar
- Shilpee Dutt laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Ketaki Bachal
- M-Lab, Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Pallavi Shirke
- M-Lab, Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, India
| | - Archisman Banerjee
- Shilpee Dutt laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Nilesh Gardi
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Abhijit Majumder
- M-Lab, Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Shilpee Dutt
- Shilpee Dutt laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India..
| |
Collapse
|
99
|
Liau LM, Ashkan K, Brem S, Campian JL, Trusheim JE, Iwamoto FM, Tran DD, Ansstas G, Cobbs CS, Heth JA, Salacz ME, D’Andre S, Aiken RD, Moshel YA, Nam JY, Pillainayagam CP, Wagner SA, Walter KA, Chaudhary R, Goldlust SA, Lee IY, Bota DA, Elinzano H, Grewal J, Lillehei K, Mikkelsen T, Walbert T, Abram S, Brenner AJ, Ewend MG, Khagi S, Lovick DS, Portnow J, Kim L, Loudon WG, Martinez NL, Thompson RC, Avigan DE, Fink KL, Geoffroy FJ, Giglio P, Gligich O, Krex D, Lindhorst SM, Lutzky J, Meisel HJ, Nadji-Ohl M, Sanchin L, Sloan A, Taylor LP, Wu JK, Dunbar EM, Etame AB, Kesari S, Mathieu D, Piccioni DE, Baskin DS, Lacroix M, May SA, New PZ, Pluard TJ, Toms SA, Tse V, Peak S, Villano JL, Battiste JD, Mulholland PJ, Pearlman ML, Petrecca K, Schulder M, Prins RM, Boynton AL, Bosch ML. Association of Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination With Extension of Survival Among Patients With Newly Diagnosed and Recurrent Glioblastoma: A Phase 3 Prospective Externally Controlled Cohort Trial. JAMA Oncol 2023; 9:112-121. [PMID: 36394838 PMCID: PMC9673026 DOI: 10.1001/jamaoncol.2022.5370] [Citation(s) in RCA: 172] [Impact Index Per Article: 172.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/27/2022] [Indexed: 11/19/2022]
Abstract
Importance Glioblastoma is the most lethal primary brain cancer. Clinical outcomes for glioblastoma remain poor, and new treatments are needed. Objective To investigate whether adding autologous tumor lysate-loaded dendritic cell vaccine (DCVax-L) to standard of care (SOC) extends survival among patients with glioblastoma. Design, Setting, and Participants This phase 3, prospective, externally controlled nonrandomized trial compared overall survival (OS) in patients with newly diagnosed glioblastoma (nGBM) and recurrent glioblastoma (rGBM) treated with DCVax-L plus SOC vs contemporaneous matched external control patients treated with SOC. This international, multicenter trial was conducted at 94 sites in 4 countries from August 2007 to November 2015. Data analysis was conducted from October 2020 to September 2021. Interventions The active treatment was DCVax-L plus SOC temozolomide. The nGBM external control patients received SOC temozolomide and placebo; the rGBM external controls received approved rGBM therapies. Main Outcomes and Measures The primary and secondary end points compared overall survival (OS) in nGBM and rGBM, respectively, with contemporaneous matched external control populations from the control groups of other formal randomized clinical trials. Results A total of 331 patients were enrolled in the trial, with 232 randomized to the DCVax-L group and 99 to the placebo group. Median OS (mOS) for the 232 patients with nGBM receiving DCVax-L was 19.3 (95% CI, 17.5-21.3) months from randomization (22.4 months from surgery) vs 16.5 (95% CI, 16.0-17.5) months from randomization in control patients (HR = 0.80; 98% CI, 0.00-0.94; P = .002). Survival at 48 months from randomization was 15.7% vs 9.9%, and at 60 months, it was 13.0% vs 5.7%. For 64 patients with rGBM receiving DCVax-L, mOS was 13.2 (95% CI, 9.7-16.8) months from relapse vs 7.8 (95% CI, 7.2-8.2) months among control patients (HR, 0.58; 98% CI, 0.00-0.76; P < .001). Survival at 24 and 30 months after recurrence was 20.7% vs 9.6% and 11.1% vs 5.1%, respectively. Survival was improved in patients with nGBM with methylated MGMT receiving DCVax-L compared with external control patients (HR, 0.74; 98% CI, 0.55-1.00; P = .03). Conclusions and Relevance In this study, adding DCVax-L to SOC resulted in clinically meaningful and statistically significant extension of survival for patients with both nGBM and rGBM compared with contemporaneous, matched external controls who received SOC alone. Trial Registration ClinicalTrials.gov Identifier: NCT00045968.
Collapse
Affiliation(s)
- Linda M. Liau
- Department of Neurosurgery, University of California, Los Angeles
| | | | - Steven Brem
- Department of Neurosurgery, Penn Brain Tumor Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jian L. Campian
- Division of Neurology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - John E. Trusheim
- Givens Brain Tumor Center, Abbott Northwestern Hospital, Minneapolis, Minnesota
| | - Fabio M. Iwamoto
- Columbia University Irving Medical Center, New York, New York
- New York-Presbyterian Hospital, New York, New York
| | - David D. Tran
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, Division of Neuro-Oncology, Lillian S. Wells Department of Neurosurgery, University of Florida College of Medicine, Gainesville
| | - George Ansstas
- Department of Neurological Surgery, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Charles S. Cobbs
- Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Medical Center, Seattle, Washington
| | - Jason A. Heth
- Taubman Medical Center, University of Michigan, Ann Arbor
| | - Michael E. Salacz
- Neuro-Oncology Program, Rutgers Cancer Institute of New Jersey, New Brunswick
| | | | - Robert D. Aiken
- Glasser Brain Tumor Center, Atlantic Healthcare, Summit, New Jersey
| | - Yaron A. Moshel
- Glasser Brain Tumor Center, Atlantic Healthcare, Summit, New Jersey
| | - Joo Y. Nam
- Department of Neurological Sciences, Rush Medical College, Chicago, Illinois
| | | | | | | | | | - Samuel A. Goldlust
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, New Jersey
| | - Ian Y. Lee
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan
| | - Daniela A. Bota
- Department of Neurology and Chao Family Comprehensive Cancer Center, University of California, Irvine
| | | | - Jai Grewal
- Long Island Brain Tumor Center at NSPC, Lake Success, New York
| | - Kevin Lillehei
- Department of Neurosurgery, University of Colorado Health Sciences Center, Boulder
| | - Tom Mikkelsen
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan
| | - Tobias Walbert
- Department of Neurosurgery, Henry Ford Health System, Detroit, Michigan
| | - Steven Abram
- Ascension St Thomas Brain and Spine Tumor Center, Howell Allen Clinic, Nashville, Tennessee
| | | | - Matthew G. Ewend
- Department of Neurosurgery, UNC School of Medicine and UNC Health, Chapel Hill, North Carolina
| | - Simon Khagi
- The Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | | | - Jana Portnow
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, California
| | - Lyndon Kim
- Division of Neuro-Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Nina L. Martinez
- Jefferson Hospital for Neurosciences, Jefferson University, Philadelphia, Pennsylvania
| | - Reid C. Thompson
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David E. Avigan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Cambridge, Massachusetts
| | - Karen L. Fink
- Baylor Scott & White Neuro-Oncology Associates, Dallas, Texas
| | | | - Pierre Giglio
- Medical University of South Carolina Neurosciences, Charleston
| | - Oleg Gligich
- Mount Sinai Medical Center, Miami Beach, Florida
| | | | - Scott M. Lindhorst
- Hollings Cancer Center, Medical University of South Carolina, Charleston
| | - Jose Lutzky
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | | | - Minou Nadji-Ohl
- Neurochirurgie Katharinenhospital, Klinikum der Landeshauptstadt Stuttgart, Stuttgart, Germany
| | | | - Andrew Sloan
- Seidman Cancer Center, University Hospitals–Cleveland Medical Center, Cleveland, Ohio
| | - Lynne P. Taylor
- Department of Neurosurgery, Tufts Medical Center, Boston, Massachusetts
| | - Julian K. Wu
- Department of Neurosurgery, Tufts Medical Center, Boston, Massachusetts
| | - Erin M. Dunbar
- Piedmont Physicians Neuro-Oncology, Piedmont Brain Tumor Center, Atlanta, Georgia
| | | | - Santosh Kesari
- Pacific Neurosciences Institute and Saint John’s Cancer Institute, Santa Monica, California
| | - David Mathieu
- Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - David S. Baskin
- Department of Neurosurgery, Houston Methodist Hospital, Houston, Texas
| | - Michel Lacroix
- Geisinger Neuroscience Institute, Danville, Pennsylvania
| | | | | | | | - Steven A. Toms
- Departments of Neurosurgery and Medicine, The Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Victor Tse
- Kaiser Permanente, Redwood City, California
| | - Scott Peak
- Kaiser Permanente, Redwood City, California
| | - John L. Villano
- University of Kentucky Markey Cancer Center, Department of Medicine, Neurosurgery, and Neurology, University of Kentucky, Lexington
| | | | | | | | - Kevin Petrecca
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Michael Schulder
- Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Uniondale, New York
| | | | | | | |
Collapse
|
100
|
González V, Brell M, Fuster J, Moratinos L, Alegre D, López S, Ibáñez J. Analyzing the role of reoperation in recurrent glioblastoma: a 15-year retrospective study in a single institution. World J Surg Oncol 2022; 20:384. [PMID: 36464682 PMCID: PMC9721080 DOI: 10.1186/s12957-022-02852-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/28/2022] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND Multiple treatment options at glioblastoma progression exist, including reintervention, reirradiation, additional systemic therapy, and novel strategies. No alternative has been proven to be superior in terms of postprogression survival (PPS). A second surgery has shown conflicting evidence in the literature regarding its prognostic impact, possibly affected by selection bias, and might benefit a sparse subset of patients with recurrent glioblastoma. The present study aims to determine the prognostic influence of salvage procedures in a cohort of patients treated in the same institution over 15 years. METHODS Three hundred and fifty patients with confirmed primary glioblastoma diagnosed and treated between 2005 and 2019 were selected. To examine the role of reoperation, we intended to create comparable groups, previously excluding all diagnostic biopsies and patients who were not actively treated after the first surgery or at disease progression. Uni- and multivariate Cox proportional hazards regression models were employed, considering reintervention as a time-fixed or time-dependent covariate. The endpoints of the study were overall survival (OS) and PPS. RESULTS At progression, 33 patients received a second surgery and 84 were treated with chemotherapy only. Clinical variables were similar among groups. OS, but not PPS, was superior in the reintervention group. Treatment modality had no impact in our multivariate Cox regression models considering OS or PPS as the endpoint. CONCLUSIONS The association of reoperation with improved prognosis in recurrent glioblastoma is unclear and may be influenced by selection bias. Regardless of our selective indications and high gross total resection rates in second procedures, we could not observe a survival advantage.
Collapse
Affiliation(s)
- Víctor González
- grid.411164.70000 0004 1796 5984Neurosurgical Department, Hospital Son Espases, Carretera de Valldemossa, 79, 07120 Palma, Illes Balears Spain
| | - Marta Brell
- grid.411164.70000 0004 1796 5984Neurosurgical Department, Hospital Son Espases, Carretera de Valldemossa, 79, 07120 Palma, Illes Balears Spain
| | - José Fuster
- grid.411164.70000 0004 1796 5984Oncology Department, Hospital Son Espases, Carretera de Valldemossa, 79, 07120 Palma, Illes Balears Spain
| | - Lesmes Moratinos
- grid.411164.70000 0004 1796 5984Neurosurgical Department, Hospital Son Espases, Carretera de Valldemossa, 79, 07120 Palma, Illes Balears Spain
| | - Daniel Alegre
- grid.411164.70000 0004 1796 5984Neurosurgical Department, Hospital Son Espases, Carretera de Valldemossa, 79, 07120 Palma, Illes Balears Spain
| | - Sofía López
- grid.411164.70000 0004 1796 5984Neurosurgical Department, Hospital Son Espases, Carretera de Valldemossa, 79, 07120 Palma, Illes Balears Spain
| | - Javier Ibáñez
- grid.411164.70000 0004 1796 5984Neurosurgical Department, Hospital Son Espases, Carretera de Valldemossa, 79, 07120 Palma, Illes Balears Spain
| |
Collapse
|