1
|
Malmberg C, Hellquist BN, Sadanandan SA, Sandström M, Wu WYY, Björkblom B, Melin B, Sjöberg RL. Antidepressant drugs and risk of developing glioma: a national registry-based case-control study and a meta-analysis. Am J Epidemiol 2024; 193:1592-1599. [PMID: 38825331 DOI: 10.1093/aje/kwae100] [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: 07/06/2023] [Revised: 04/25/2024] [Accepted: 05/28/2024] [Indexed: 06/04/2024] Open
Abstract
Whether use of antidepressants is related to the risk of developing lower-grade (WHO grades 2-3) and higher-grade (WHO grade 4) glioma was investigated in this study. A registry-based case-control study was performed with 1283 glioma case patients and 6400 age-, sex-, and geographically matched control participants who were diagnosed in Sweden during 2009-2013. Conditional logistic regression was used to analyze whether selective serotonin reuptake inhibitors (SSRIs) or non-SSRIs were associated with the risk of developing lower- or higher-grade glioma in the study population. Our results show that use of antidepressant medication was not associated with the risk of developing glioma. We also performed a meta-analysis in which the data set from the present study was combined with results from 2 previous epidemiologic studies to answer the same questions. The meta-analysis showed a modest risk reduction of developing glioma in relation to antidepressant treatment (odds ratio = 0.90; 95% CI, 0.83-0.97) when all glioma subgroups and all forms of antidepressant medications were combined. In conclusion, it remains possible that antidepressants may have common monoaminergic mechanism(s) that reduce the risk of developing glioma.
Collapse
Affiliation(s)
- Charlotte Malmberg
- Department of Clinical Sciences, Neurosciences, Umeå University, S-901 85 Umeå, Sweden
| | | | | | - Maria Sandström
- Department of Radiation Sciences, Oncology, Umeå University, S-901 87 Umeå, Sweden
| | - Wendy Yi-Ying Wu
- Department of Radiation Sciences, Oncology, Umeå University, S-901 87 Umeå, Sweden
| | - Benny Björkblom
- Department of Chemistry, Umeå University, S-901 87 Umeå, Sweden
| | - Beatrice Melin
- Department of Radiation Sciences, Oncology, Umeå University, S-901 87 Umeå, Sweden
| | - Rickard L Sjöberg
- Department of Clinical Sciences, Neurosciences, Umeå University, S-901 85 Umeå, Sweden
| |
Collapse
|
2
|
Gonzales CN, Negussie MB, Krishna S, Ambati VS, Hervey-Jumper SL. Malignant glioma remodeling of neuronal circuits: therapeutic opportunities and repurposing of antiepileptic drugs. Trends Cancer 2024:S2405-8033(24)00192-4. [PMID: 39327186 DOI: 10.1016/j.trecan.2024.09.003] [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: 06/04/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024]
Abstract
Tumor-associated epilepsy is the most common presenting symptom in patients diagnosed with diffuse gliomas. Recent evidence illustrates the requirement of synaptic activity to drive glioma proliferation and invasion. Class 1, 2, and 3 evidence is limited regarding the use of antiepileptic drugs (AEDs) as antitumor therapy in combination with chemotherapy. Furthermore, no central mechanism has emerged as the most targetable. The optimal timing of AED regimen remains unknown. Targeting aberrant neuronal activity is a promising avenue for glioma treatment. Clinical biomarkers may aid in identifying patients most likely to benefit from AEDs. Quality evidence is needed to guide treatment decisions.
Collapse
Affiliation(s)
- Cesar Nava Gonzales
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Mikias B Negussie
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Saritha Krishna
- Department of Neurological Surgery, University of California, San Francisco, CA, USA; Weill Institute of Neurosciences, University of California, San Francisco, CA, USA
| | - Vardhaan S Ambati
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Shawn L Hervey-Jumper
- Department of Neurological Surgery, University of California, San Francisco, CA, USA; Weill Institute of Neurosciences, University of California, San Francisco, CA, USA.
| |
Collapse
|
3
|
Lee S, Weiss T, Bühler M, Mena J, Lottenbach Z, Wegmann R, Sun M, Bihl M, Augustynek B, Baumann SP, Goetze S, van Drogen A, Pedrioli PGA, Penton D, Festl Y, Buck A, Kirschenbaum D, Zeitlberger AM, Neidert MC, Vasella F, Rushing EJ, Wollscheid B, Hediger MA, Weller M, Snijder B. High-throughput identification of repurposable neuroactive drugs with potent anti-glioblastoma activity. Nat Med 2024:10.1038/s41591-024-03224-y. [PMID: 39304781 DOI: 10.1038/s41591-024-03224-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/31/2024] [Indexed: 09/22/2024]
Abstract
Glioblastoma, the most aggressive primary brain cancer, has a dismal prognosis, yet systemic treatment is limited to DNA-alkylating chemotherapies. New therapeutic strategies may emerge from exploring neurodevelopmental and neurophysiological vulnerabilities of glioblastoma. To this end, we systematically screened repurposable neuroactive drugs in glioblastoma patient surgery material using a clinically concordant and single-cell resolved platform. Profiling more than 2,500 ex vivo drug responses across 27 patients and 132 drugs identified class-diverse neuroactive drugs with potent anti-glioblastoma efficacy that were validated across model systems. Interpretable molecular machine learning of drug-target networks revealed neuroactive convergence on AP-1/BTG-driven glioblastoma suppression, enabling expanded in silico screening of more than 1 million compounds with high patient validation accuracy. Deep multimodal profiling confirmed Ca2+-driven AP-1/BTG-pathway induction as a neuro-oncological glioblastoma vulnerability, epitomized by the anti-depressant vortioxetine synergizing with current standard-of-care chemotherapies in vivo. These findings establish an actionable framework for glioblastoma treatment rooted in its neural etiology.
Collapse
Affiliation(s)
- Sohyon Lee
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Marcel Bühler
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Julien Mena
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Zuzanna Lottenbach
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Rebekka Wegmann
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Miaomiao Sun
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Michel Bihl
- Institute of Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Bartłomiej Augustynek
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Bern, Switzerland
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Sven P Baumann
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Bern, Switzerland
| | - Sandra Goetze
- Department of Health Sciences and Technology, Institute of Translational Medicine (ITM), ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Zurich, Switzerland
| | - Audrey van Drogen
- Department of Health Sciences and Technology, Institute of Translational Medicine (ITM), ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Zurich, Switzerland
| | - Patrick G A Pedrioli
- Department of Health Sciences and Technology, Institute of Translational Medicine (ITM), ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Zurich, Switzerland
| | - David Penton
- Electrophysiology Facility, University of Zurich, Zurich, Switzerland
| | - Yasmin Festl
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Alicia Buck
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Daniel Kirschenbaum
- Department of Neuropathology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Anna M Zeitlberger
- Department of Neurosurgery, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Marian C Neidert
- Department of Neurosurgery, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Flavio Vasella
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Elisabeth J Rushing
- Department of Neuropathology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Health Sciences and Technology, Institute of Translational Medicine (ITM), ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- ETH PHRT Swiss Multi-Omics Center (SMOC), Zurich, Switzerland
| | - Matthias A Hediger
- Membrane Transport Discovery Lab, Department of Nephrology and Hypertension and Department of Biomedical Research, Inselspital, University of Bern, Bern, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Berend Snijder
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Comprehensive Cancer Center Zurich, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
4
|
Speranza L, Filiz KD, Lippiello P, Ferraro MG, Pascarella S, Miniaci MC, Volpicelli F. Enduring Neurobiological Consequences of Early-Life Stress: Insights from Rodent Behavioral Paradigms. Biomedicines 2024; 12:1978. [PMID: 39335492 PMCID: PMC11429222 DOI: 10.3390/biomedicines12091978] [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: 06/17/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Stress profoundly affects physical and mental health, particularly when experienced early in life. Early-life stress (ELS) encompasses adverse childhood experiences such as abuse, neglect, violence, or chronic poverty. These stressors can induce long-lasting changes in brain structure and function, impacting areas involved in emotion regulation, cognition, and stress response. Consequently, individuals exposed to high levels of ELS are at an increased risk for mental health disorders like depression, anxiety, and post-traumatic stress disorders, as well as physical health issues, including metabolic disorders, cardiovascular disease, and cancer. This review explores the biological and psychological consequences of early-life adversity paradigms in rodents, such as maternal separation or deprivation and limited bedding or nesting. The study of these experimental models have revealed that the organism's response to ELS is complex, involving genetic and epigenetic mechanisms, and is associated with the dysregulation of physiological systems like the nervous, neuroendocrine, and immune systems, in a sex-dependent fashion. Understanding the impact of ELS is crucial for developing effective interventions and preventive strategies in humans exposed to stressful or traumatic experiences in childhood.
Collapse
Affiliation(s)
- Luisa Speranza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Kardelen Dalim Filiz
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Pellegrino Lippiello
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Silvia Pascarella
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Maria Concetta Miniaci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| |
Collapse
|
5
|
Zheng G, Zeng G, Wei D. The role of NPC2 gene in glioma was investigated based on bioinformatics analysis. Sci Rep 2024; 14:19134. [PMID: 39160329 PMCID: PMC11333723 DOI: 10.1038/s41598-024-70221-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/14/2024] [Indexed: 08/21/2024] Open
Abstract
Glioblastoma (GBM) is one of the most malignant primary brain tumors in adults. The NPC2 gene (Niemann-Pick type C intracellular cholesterol transporter 2) is a protein-coding gene with a lipid recognition domain. The NPC2 gene was found to be significantly increased in gliomas (LGG and GBM), and it is now thought to be a risk factor. COX analysis demonstrated that NPC2 was a significant risk factor for glioma. Functional enrichment analysis of genes that were differentially expressed between high and low expression groups revealed that genes were primarily enriched in the regulation of trans-synaptic signaling, Retrograde endocannabinoid signaling and other pathways. According to the findings of the immunoinfiltration investigation, the NPC2 gene and macrophage, DC, etc. have a strong positive association. In addition, patients with high NPC2 expression had higher levels of immune cell expression. Medication sensitivity research revealed that NPC2's differential expression had some bearing on patients' medication sensitivity. There was a strong correlation between the prognosis of glioma patients and the gene sets NUDT19 and NUME. In brief, the NPC2 gene was identified to be a possible biomarker of glioma, and preliminary analysis was done on the role of the NPC2 gene in immunological microenvironment of glioma.
Collapse
Affiliation(s)
- Guangwei Zheng
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Neurosurgery, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Guangming Zeng
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Neurosurgery, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - De Wei
- Department of Neurosurgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.
- Department of Neurosurgery, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
6
|
Zhang H, Hong Y, Wu T, Ben E, Li S, Hu L, Xie T. Role of gut microbiota in regulating immune checkpoint inhibitor therapy for glioblastoma. Front Immunol 2024; 15:1401967. [PMID: 38915399 PMCID: PMC11194316 DOI: 10.3389/fimmu.2024.1401967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/28/2024] [Indexed: 06/26/2024] Open
Abstract
Glioblastoma (GBM) is a highly malignant, invasive, and poorly prognosed brain tumor. Unfortunately, active comprehensive treatment does not significantly prolong patient survival. With the deepening of research, it has been found that gut microbiota plays a certain role in GBM, and can directly or indirectly affect the efficacy of immune checkpoint inhibitors (ICIs) in various ways. (1) The metabolites produced by gut microbiota directly affect the host's immune homeostasis, and these metabolites can affect the function and distribution of immune cells, promote or inhibit inflammatory responses, affect the phenotype, angiogenesis, inflammatory response, and immune cell infiltration of GBM cells, thereby affecting the effectiveness of ICIs. (2) Some members of the gut microbiota may reverse T cell function inhibition, increase T cell anti-tumor activity, and ultimately improve the efficacy of ICIs by targeting specific immunosuppressive metabolites and cytokines. (3) Some members of the gut microbiota directly participate in the metabolic process of drugs, which can degrade, transform, or produce metabolites, affecting the effective concentration and bioavailability of drugs. Optimizing the structure of the gut microbiota may help improve the efficacy of ICIs. (4) The gut microbiota can also regulate immune cell function and inflammatory status in the brain through gut brain axis communication, indirectly affecting the progression of GBM and the therapeutic response to ICIs. (5) Given the importance of gut microbiota for ICI therapy, researchers have begun exploring the use of fecal microbiota transplantation (FMT) to transplant healthy or optimized gut microbiota to GBM patients, in order to improve their immune status and enhance their response to ICI therapy. Preliminary studies suggest that FMT may enhance the efficacy of ICI therapy in some patients. In summary, gut microbiota plays a crucial role in regulating ICIs in GBM, and with a deeper understanding of the relationship between gut microbiota and tumor immunity, it is expected to develop more precise and effective personalized ICI therapy strategies for GBM, in order to improve patient prognosis.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Radiation Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Hong
- Department of Radiation Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tingting Wu
- Department of Health Management, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Eyi Ben
- Department of Oncology, Yidu People’s Hospital, Yichang, Hubei, China
| | - Shuai Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liu Hu
- Department of Radiation Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Xie
- Department of Radiation Oncology, Hubei Cancer Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
7
|
Chang YC, Chan MH, Li CH, Chen CL, Tsai WC, Hsiao M. PPAR-γ agonists reactivate the ALDOC-NR2F1 axis to enhance sensitivity to temozolomide and suppress glioblastoma progression. Cell Commun Signal 2024; 22:266. [PMID: 38741139 PMCID: PMC11089732 DOI: 10.1186/s12964-024-01645-3] [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: 12/07/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Glioblastoma (GBM) is a type of brain cancer categorized as a high-grade glioma. GBM is characterized by limited treatment options, low patient survival rates, and abnormal serotonin metabolism. Previous studies have investigated the tumor suppressor function of aldolase C (ALDOC), a glycolytic enzyme in GBM. However, it is unclear how ALDOC regulates production of serotonin and its associated receptors, HTRs. In this study, we analyzed ALDOC mRNA levels and methylation status using sequencing data and in silico datasets. Furthermore, we investigated pathways, phenotypes, and drug effects using cell and mouse models. Our results suggest that loss of ALDOC function in GBM promotes tumor cell invasion and migration. We observed that hypermethylation, which results in loss of ALDOC expression, is associated with serotonin hypersecretion and the inhibition of PPAR-γ signaling. Using several omics datasets, we present evidence that ALDOC regulates serotonin levels and safeguards PPAR-γ against serotonin metabolism mediated by 5-HT, which leads to a reduction in PPAR-γ expression. PPAR-γ activation inhibits serotonin release by HTR and diminishes GBM tumor growth in our cellular and animal models. Importantly, research has demonstrated that PPAR-γ agonists prolong animal survival rates and increase the efficacy of temozolomide in an orthotopic brain model of GBM. The relationship and function of the ALDOC-PPAR-γ axis could serve as a potential prognostic indicator. Furthermore, PPAR-γ agonists offer a new treatment alternative for glioblastoma multiforme (GBM).
Collapse
Affiliation(s)
- Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| | - Ming-Hsien Chan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Chien-Hsiu Li
- Department of Urology, Shuang Ho Hospital, Taipei Medical University, New Taipei, 235, Taiwan
| | - Chi-Long Chen
- Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, 110, Taiwan
- Department of Pathology, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| |
Collapse
|
8
|
Jackson ER, Persson ML, Fish CJ, Findlay IJ, Mueller S, Nazarian J, Hulleman E, van der Lugt J, Duchatel RJ, Dun MD. A review of current therapeutics targeting the mitochondrial protease ClpP in diffuse midline glioma, H3 K27-altered. Neuro Oncol 2024; 26:S136-S154. [PMID: 37589388 PMCID: PMC11066926 DOI: 10.1093/neuonc/noad144] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Indexed: 08/18/2023] Open
Abstract
Diffuse midline gliomas (DMGs) are devastating pediatric brain tumors recognized as the leading cause of cancer-related death in children. DMGs are high-grade gliomas (HGGs) diagnosed along the brain's midline. Euchromatin is the hallmark feature of DMG, caused by global hypomethylation of H3K27 either through point mutations in histone H3 genes (H3K27M), or by overexpression of the enhancer of zeste homolog inhibitory protein. In a clinical trial for adults with progressive HGGs, a 22-year-old patient with a thalamic DMG, H3 K27-altered, showed a remarkable clinical and radiological response to dordaviprone (ONC201). This response in an H3 K27-altered HGG patient, coupled with the lack of response of patients harboring wildtype-H3 tumors, has increased the clinical interest in dordaviprone for the treatment of DMG. Additional reports of clinical benefit have emerged, but research defining mechanisms of action (MOA) fall behind dordaviprone's clinical use, with biomarkers of response unresolved. Here, we summarize dordaviprone's safety, interrogate its preclinical MOA identifying the mitochondrial protease "ClpP" as a biomarker of response, and discuss other ClpP agonists, expanding the arsenal of potential weapons in the fight against DMG. Finally, we discuss combination strategies including ClpP agonists, and their immunomodulatory effects suggestive of a role for the tumor microenvironment in DMG patient response.
Collapse
Affiliation(s)
- Evangeline R Jackson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Mika L Persson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Cameron J Fish
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Izac J Findlay
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Sabine Mueller
- DIPG/DMG Center Zurich, University Children’s Hospital Zürich, Zurich, Switzerland
- Department of Neurology, Neurosurgery and Pediatric, UCSF, San Francisco, California, USA
| | - Javad Nazarian
- DIPG/DMG Center Zurich, University Children’s Hospital Zürich, Zurich, Switzerland
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
- The George Washington University, School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands, Utrecht, Netherlands
| | - Jasper van der Lugt
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands, Utrecht, Netherlands
| | - Ryan J Duchatel
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
- Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
9
|
Edström S, Hellquist BN, Sandström M, Sadanandan SA, Björkblom B, Melin B, Sjöberg RL. Antidepressants and survival in glioma-A registry-based retrospective cohort study. Neurooncol Pract 2024; 11:125-131. [PMID: 38496917 PMCID: PMC10940821 DOI: 10.1093/nop/npad057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Abstract
Background Depression and treatment with antidepressant medication is common in patients with malignant glioma. However, the extent to which antidepressants may affect the disease is not fully understood. Therefore, the purpose of the present study was to investigate possible associations between treatment with antidepressant medication and survival in glioma patients. Methods We performed a registry-based cohort study including 1231 patients with malignant glioma (WHO grades 2, 3, and 4) having undergone surgery, and 6400 matched controls without glioma. All data were extracted from the RISK North database, which contains information from multiple national population-based registries in Sweden. Results Treatment with antidepressants is more common in patients with malignant glioma (27%), compared to controls (16%), P < .001. Treatment with antidepressants after surgery for glioma was significantly associated with poorer survival. These effects were observed both for selective serotonin reuptake inhibitors (SSRIs) and non-SSRIs. In grade 4 glioma, SSRI treatment was associated with a hazard ratio (HR) of 3.32 (95% CI 2.69-4.10, P < .001), and non-SSRI treatment a HR of 3.54 (95% CI 2.52-4.99, P < .001), compared to glioma patients without antidepressants. In grade 2-3 glioma, the HR for SSRI treatment was 3.26 (95% CI 2.19-4.85, P < .001), and for non-SSRI treatment was 7.71 (95% CI 4.22-14.12, P < .001). Conclusions Our results demonstrate a negative association between antidepressant medication and survival in glioma. Further research will be needed to clarify causation.
Collapse
Affiliation(s)
- Sonja Edström
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | | | - Maria Sandström
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | | | | | - Beatrice Melin
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Rickard L Sjöberg
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| |
Collapse
|
10
|
Tripathy S, Singh S, Banerjee M, Modi DR, Prakash A. Coagulation proteases and neurotransmitters in pathogenicity of glioblastoma multiforme. Int J Neurosci 2024; 134:398-408. [PMID: 35896309 DOI: 10.1080/00207454.2022.2107514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 07/10/2022] [Accepted: 07/15/2022] [Indexed: 10/16/2022]
Abstract
Glioblastoma is an aggressive type of cancer that begins in cells called astrocytes that support nerve cells that can occur in the brain or spinal cord. It can form in the brain or spinal cord. Despite the variety of modern therapies against GBM, it is still a deadly disease. Patients usually have a median survival of approximately 14 to 15 months from the diagnosis. Glioblastoma is also known as glioblastoma multiforme. The pathogenesis contributing to the proliferation and metastasis of cancer involves aberrations of multiple signalling pathways through multiple genetic mutations and altered gene expression. The coagulant factors like thrombin and tissue factor play a noteworthy role in cancer invasion. They are produced in the microenvironment of glioma through activation of protease-activated receptors (PARs) which are activated by coagulation proteases. PARs are members of family G-protein-coupled receptors (GPCRs) that are activated by coagulation proteases. These components play a key role in tumour cell angiogenesis, migration, invasion, and interactions with host vascular cells. Further, the release of neurotransmitters is also found to regulate malignancy in gliomas. Exploration of the interplay between malignant neural circuitry with the normal conditions is also decisive in finding effective therapies for these apparently invasive tumours. The present review discusses the molecular classification of gliomas, activation of PARs by coagulation protease, and its role in metastasis of gliomas. Further, the differential involvement of neurotransmitters in the pathogenesis of gliomas has also been discussed. Targeting these molecules may present a potential therapeutic approach for the treatment of gliomas.
Collapse
Affiliation(s)
- Sukanya Tripathy
- Molecular & Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Sanjay Singh
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Monisha Banerjee
- Molecular & Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
| | - Dinesh Raj Modi
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Anand Prakash
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| |
Collapse
|
11
|
Xu X, Han Y, Zhang B, Ren Q, Ma J, Liu S. Understanding immune microenvironment alterations in the brain to improve the diagnosis and treatment of diverse brain diseases. Cell Commun Signal 2024; 22:132. [PMID: 38368403 PMCID: PMC10874090 DOI: 10.1186/s12964-024-01509-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: 09/25/2023] [Accepted: 02/01/2024] [Indexed: 02/19/2024] Open
Abstract
Abnormal inflammatory states in the brain are associated with a variety of brain diseases. The dynamic changes in the number and function of immune cells in cerebrospinal fluid (CSF) are advantageous for the early prediction and diagnosis of immune diseases affecting the brain. The aggregated factors and cells in inflamed CSF may represent candidate targets for therapy. The physiological barriers in the brain, such as the blood‒brain barrier (BBB), establish a stable environment for the distribution of resident immune cells. However, the underlying mechanism by which peripheral immune cells migrate into the brain and their role in maintaining immune homeostasis in CSF are still unclear. To advance our understanding of the causal link between brain diseases and immune cell status, we investigated the characteristics of immune cell changes in CSF and the molecular mechanisms involved in common brain diseases. Furthermore, we summarized the diagnostic and treatment methods for brain diseases in which immune cells and related cytokines in CSF are used as targets. Further investigations of the new immune cell subtypes and their contributions to the development of brain diseases are needed to improve diagnostic specificity and therapy.
Collapse
Affiliation(s)
- Xiaotong Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yi Han
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People's Republic of China.
| | - Binlong Zhang
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People's Republic of China
| | - Quanzhong Ren
- JST Sarcopenia Research Centre, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Juan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, People's Republic of China.
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People's Republic of China
| |
Collapse
|
12
|
Zhang L, Gao H, Li X, Yu F, Li P. The important regulatory roles of circRNA‑encoded proteins or peptides in cancer pathogenesis (Review). Int J Oncol 2024; 64:19. [PMID: 38186313 PMCID: PMC10783939 DOI: 10.3892/ijo.2023.5607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/13/2023] [Indexed: 01/09/2024] Open
Abstract
Circular RNAs (circRNAs) represent a class of RNA molecules characterized by their covalently closed structures. There are three types of circRNAs, namely exonic circRNAs, exon‑intron circRNAs and circular intronic RNAs. To date, four distinct mechanisms have been unveiled through which circRNAs exert their functional influence, including serving as microRNA (miRNA) sponges, interacting with RNA binding proteins (RBPs), modulating parental gene transcription and acting as templates for translation. Of note, among these mechanisms, the miRNA/RBP sponge function has been the most investigated one. Recent research has uncovered the presence of various proteins or peptides encoded by circRNA. CircRNAs are translated independent of the 5' cap and 3' polyA tail, which are typical elements for linear RNA translation. Some unique elements, such as internal ribosome entry sites and N‑methyladenosine modifications, facilitate the initiation of translation. These circRNA‑encoded proteins or peptides participate in diverse signalling pathways and act as important regulators in carcinogenesis by influencing cell proliferation, migration, apoptosis and other key processes. Consequently, circRNA‑encoded proteins or peptides have great potential as therapeutic targets for anticancer drugs. The present comprehensive review aimed to systematically summarize the current understanding of circRNA‑encoded proteins or peptides and to unveil their roles in carcinogenesis.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Huijuan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Xin Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Fei Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong 266021, P.R. China
| |
Collapse
|
13
|
Xue Z, Zhang Y, Zhao R, Liu X, Grützmann K, Klink B, Zhang X, Wang S, Zhao W, Sun Y, Han M, Wang X, Hu Y, Liu X, Yang N, Qiu C, Li W, Huang B, Li X, Bjerkvig R, Wang J, Zhou W. The dopamine receptor D1 inhibitor, SKF83566, suppresses GBM stemness and invasion through the DRD1-c-Myc-UHRF1 interactions. J Exp Clin Cancer Res 2024; 43:25. [PMID: 38246990 PMCID: PMC10801958 DOI: 10.1186/s13046-024-02947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/06/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Extensive local invasion of glioblastoma (GBM) cells within the central nervous system (CNS) is one factor that severely limits current treatments. The aim of this study was to uncover genes involved in the invasion process, which could also serve as therapeutic targets. For the isolation of invasive GBM cells from non-invasive cells, we used a three-dimensional organotypic co-culture system where glioma stem cell (GSC) spheres were confronted with brain organoids (BOs). Using ultra-low input RNA sequencing (ui-RNA Seq), an invasive gene signature was obtained that was exploited in a therapeutic context. METHODS GFP-labeled tumor cells were sorted from invasive and non-invasive regions within co-cultures. Ui-RNA sequencing analysis was performed to find a gene cluster up-regulated in the invasive compartment. This gene cluster was further analyzed using the Connectivity MAP (CMap) database. This led to the identification of SKF83566, an antagonist of the D1 dopamine receptor (DRD1), as a candidate therapeutic molecule. Knockdown and overexpression experiments were performed to find molecular pathways responsible for the therapeutic effects of SKF83566. Finally, the effects of SKF83566 were validated in orthotopic xenograft models in vivo. RESULTS Ui-RNA seq analysis of three GSC cell models (P3, BG5 and BG7) yielded a set of 27 differentially expressed genes between invasive and non-invasive cells. Using CMap analysis, SKF83566 was identified as a selective inhibitor targeting both DRD1 and DRD5. In vitro studies demonstrated that SKF83566 inhibited tumor cell proliferation, GSC sphere formation, and invasion. RNA sequencing analysis of SKF83566-treated P3, BG5, BG7, and control cell populations yielded a total of 32 differentially expressed genes, that were predicted to be regulated by c-Myc. Of these, the UHRF1 gene emerged as the most downregulated gene following treatment, and ChIP experiments revealed that c-Myc binds to its promoter region. Finally, SKF83566, or stable DRD1 knockdown, inhibited the growth of orthotopic GSC (BG5) derived xenografts in nude mice. CONCLUSIONS DRD1 contributes to GBM invasion and progression by regulating c-Myc entry into the nucleus that affects the transcription of the UHRF1 gene. SKF83566 inhibits the transmembrane protein DRD1, and as such represents a candidate small therapeutic molecule for GBMs.
Collapse
Affiliation(s)
- Zhiyi Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Ruiqi Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xiaofei Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Konrad Grützmann
- Core Unit for Molecular Tumour Diagnostics (CMTD), National Center for Tumour Diseases (NCT) Dresden, Dresden, Germany
- Institute for Medical Informatics and Biometry, Medical Faculty, TU Dresden, Dresden, Germany
| | - Barbara Klink
- Department of Genetics, Laboratoire National de Santé, Dudelange, Luxembourg
| | - Xun Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Shuai Wang
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Wenbo Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Yanfei Sun
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xu Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaotian Hu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xuemeng Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Ning Yang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Chen Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Rolf Bjerkvig
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen, 5009, Norway
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine, Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen, 5009, Norway.
| | - Wenjing Zhou
- Department of Blood Transfusion, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
14
|
Pace A, Lombardi G, Villani V, Benincasa D, Abbruzzese C, Cestonaro I, Corrà M, Padovan M, Cerretti G, Caccese M, Silvani A, Gaviani P, Giannarelli D, Ciliberto G, Paggi MG. Efficacy and safety of chlorpromazine as an adjuvant therapy for glioblastoma in patients with unmethylated MGMT gene promoter: RACTAC, a phase II multicenter trial. Front Oncol 2023; 13:1320710. [PMID: 38162492 PMCID: PMC10755935 DOI: 10.3389/fonc.2023.1320710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Drug repurposing is a promising strategy to develop new treatments for glioblastoma. In this phase II clinical trial, we evaluated the addition of chlorpromazine to temozolomide in the adjuvant phase of the standard first-line therapeutic protocol in patients with unmethylated MGMT gene promoter. Methods This was a multicenter phase II single-arm clinical trial. The experimental procedure involved the combination of CPZ with standard treatment with TMZ in the adjuvant phase of the Stupp protocol in newly-diagnosed GBM patients carrying an unmethylated MGMT gene promoter. Progression-free survival was the primary endpoint. Secondary endpoints were overall survival and toxicity. Results Forty-one patients were evaluated. Twenty patients (48.7%) completed 6 cycles of treatment with TMZ+CPZ. At 6 months, 27 patients (65.8%) were without progression, achieving the primary endpoint. Median PFS was 8.0 months (95% CI: 7.0-9.0). Median OS was 15.0 months (95% CI: 13.1-16.9). Adverse events led to reduction or interruption of CPZ dosage in 4 patients (9.7%). Discussion The addition of CPZ to standard TMZ in the first-line treatment of GBM patients with unmethylated MGMT gene promoter was safe and led to a longer PFS than expected in this population of patients. These findings provide proof-of-concept for the potential of adding CPZ to standard TMZ treatment in GBM patients with unmethylated MGMT gene promoter. Clinical trial registration https://clinicaltrials.gov/study/NCT04224441, identifier NCT04224441.
Collapse
Affiliation(s)
- Andrea Pace
- IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | | | | | | | | | | | - Martina Corrà
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Marta Padovan
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Mario Caccese
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | | | | | | | - Marco G. Paggi
- IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
15
|
Shi L, Chen H, Chen K, Zhong C, Song C, Huang Y, Wang T, Chen L, Li C, Huang A, Qi S, Li H, Lu Y. The DRD2 Antagonist Haloperidol Mediates Autophagy-Induced Ferroptosis to Increase Temozolomide Sensitivity by Promoting Endoplasmic Reticulum Stress in Glioblastoma. Clin Cancer Res 2023; 29:3172-3188. [PMID: 37249604 DOI: 10.1158/1078-0432.ccr-22-3971] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/23/2023] [Accepted: 05/25/2023] [Indexed: 05/31/2023]
Abstract
PURPOSE Temozolomide resistance remains a major obstacle in the treatment of glioblastoma (GBM). The combination of temozolomide with another agent could offer an improved treatment option if it could overcome chemoresistance and prevent side effects. Here, we determined the critical drug that cause ferroptosis in GBM cells and elucidated the possible mechanism by which drug combination overcomes chemoresistance. EXPERIMENTAL DESIGN Haloperidol/temozolomide synergism was assessed in GBM cell lines with different dopamine D2 receptor (DRD2) expression in vitro and in vivo. Inhibitors of ferroptosis, autophagy, endoplasmic reticulum (ER) stress and cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) were used to validate the specific mechanisms by which haloperidol and temozolomide induce ferroptosis in GBM cells. RESULTS In the present work, we demonstrate that the DRD2 level is increased by temozolomide in a time-dependent manner and is inversely correlated with temozolomide sensitivity in GBM. The DRD2 antagonist haloperidol, a butylbenzene antipsychotic, markedly induces ferroptosis and effectively enhances temozolomide efficacy in vivo and in vitro. Mechanistically, haloperidol suppressed the effect of temozolomide on cAMP by antagonizing DRD2 receptor activity, and the increases in cAMP/PKA triggered ER stress, which led to autophagy and ferroptosis. Furthermore, elevated autophagy mediates downregulation of FTH1 expression at the posttranslational level in an autophagy-dependent manner and ultimately leads to ferroptosis. CONCLUSIONS Our results provide experimental evidence for repurposing haloperidol as an effective adjunct therapy to inhibit adaptive temozolomide resistance to enhance the efficacy of chemoradiotherapy in GBM, a strategy that may have broad prospects for clinical application.
Collapse
Affiliation(s)
- Linyong Shi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hanning Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kunxiang Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chengzong Zhong
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chong Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Neurosurgery, The Central Hospital of Dalian University of Technology, Dalian, China
| | - Yifeng Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tong Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chiyang Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Annie Huang
- Brain Tumor Research Center, SickKids Hospital, Toronto, Canada
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Guangzhou, China
- Institute of Brain Disease, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Hong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Guangzhou, China
- Institute of Brain Disease, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yuntao Lu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Glioma Center, Guangzhou, China
- Institute of Brain Disease, Nanfang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
16
|
Lines CL, McGrath MJ, Dorwart T, Conn CS. The integrated stress response in cancer progression: a force for plasticity and resistance. Front Oncol 2023; 13:1206561. [PMID: 37601686 PMCID: PMC10435748 DOI: 10.3389/fonc.2023.1206561] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/07/2023] [Indexed: 08/22/2023] Open
Abstract
During their quest for growth, adaptation, and survival, cancer cells create a favorable environment through the manipulation of normal cellular mechanisms. They increase anabolic processes, including protein synthesis, to facilitate uncontrolled proliferation and deplete the tumor microenvironment of resources. As a dynamic adaptation to the self-imposed oncogenic stress, cancer cells promptly hijack translational control to alter gene expression. Rewiring the cellular proteome shifts the phenotypic balance between growth and adaptation to promote therapeutic resistance and cancer cell survival. The integrated stress response (ISR) is a key translational program activated by oncogenic stress that is utilized to fine-tune protein synthesis and adjust to environmental barriers. Here, we focus on the role of ISR signaling for driving cancer progression. We highlight mechanisms of regulation for distinct mRNA translation downstream of the ISR, expand on oncogenic signaling utilizing the ISR in response to environmental stresses, and pinpoint the impact this has for cancer cell plasticity during resistance to therapy. There is an ongoing need for innovative drug targets in cancer treatment, and modulating ISR activity may provide a unique avenue for clinical benefit.
Collapse
Affiliation(s)
| | | | | | - Crystal S. Conn
- Department of Radiation Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
17
|
Tanrıkulu B, Yaşar AH, Canpolat C, Çorapçıoğlu F, Tezcanli E, Abacioglu U, Danyeli AE, Özek MM. Preliminary findings of German-sourced ONC201 treatment in H3K27 altered pediatric pontine diffuse midline gliomas. J Neurooncol 2023; 163:565-575. [PMID: 37402093 DOI: 10.1007/s11060-023-04347-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/16/2023] [Indexed: 07/05/2023]
Abstract
PURPOSE H3K27 altered pediatric pontine diffuse midline gliomas (pDMG) have a poor prognosis, and conventional treatments offer limited benefits. However, recent advancements in molecular evaluations and targeted therapies have shown promise. The aim of this retrospective analysis was to evaluate the effectiveness of German-sourced ONC201, a selective antagonist of dopamine receptor DRD2, for the treatment of pediatric H3K27 altered pDMGs. METHODS Pediatric patients with H3K27 altered pDMG treated between January 2016 and July 2022 were included in this retrospective analysis. Tissue samples were acquired from all patients via stereotactic biopsy for immunohistochemistry and molecular profiling. All patients received radiation treatment with concurrent temozolomide, and those who could acquire GsONC201 received it as a single agent until progression. Patients who could not obtain GsONC201 received other chemotherapy protocols. RESULTS Among 27 patients with a median age of 5.6 years old (range 3.4-17.9), 18 received GsONC201. During the follow-up period, 16 patients (59.3%) had progression, although not statistically significant, the incidence of progression tended to be lower in the GsONC201 group. The median overall survival (OS) of the GsONC201 group was considerably longer than of the non-GsONC201 group (19.9 vs. 10.9 months). Only two patients receiving GsONC201 experienced fatigue as a side effect. 4 out of 18 patients in the GsONC201 group underwent reirradiation after progression. CONCLUSION In conclusion, this study suggests that GsONC201 may improve OS in pediatric H3K27-altered pDMG patients without significant side effects. However, caution is warranted due to retrospective design and biases, highlighting the need for further randomized clinical studies to validate these findings.
Collapse
Affiliation(s)
- Bahattin Tanrıkulu
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Acibadem University School of Medicine, Istanbul, Turkey.
- Acibadem Altunizade Hospital, Yurtcan sk No. 1, Üsküdar/Istanbul, Turkey.
| | - Ahmet Harun Yaşar
- Department of Neurosurgery, Acibadem University School of Medicine, Istanbul, Turkey
| | - Cengiz Canpolat
- Division of Hematology and Oncology, Department of Pediatrics, Acibadem University School of Medicine, Istanbul, Turkey
| | - Funda Çorapçıoğlu
- Division of Hematology and Oncology, Department of Pediatrics, Acibadem Maslak Hospital, Istanbul, Turkey
| | - Evrim Tezcanli
- Department of Radiation Oncology, Acibadem University School of Medicine, Istanbul, Turkey
| | - Ufuk Abacioglu
- Department of Radiation Oncology, Acibadem University School of Medicine, Istanbul, Turkey
| | - Ayça Erşen Danyeli
- Division of Neuropathology, Department of Pathology, Acibadem University School of Medicine, Istanbul, Turkey
| | - M Memet Özek
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Acibadem University School of Medicine, Istanbul, Turkey
| |
Collapse
|
18
|
Jeon HM, Oh YT, Shin YJ, Chang N, Kim D, Woo D, Yeup Y, Joo KM, Jo H, Yang H, Lee JK, Kang W, Sa J, Lee WJ, Hale J, Lathia JD, Purow B, Park MJ, Park JB, Nam DH, Lee J. Dopamine receptor D2 regulates glioblastoma survival and death through MET and death receptor 4/5. Neoplasia 2023; 39:100894. [PMID: 36972629 PMCID: PMC10066565 DOI: 10.1016/j.neo.2023.100894] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/28/2023] [Indexed: 03/29/2023]
Abstract
Recent studies indicate that signaling molecules traditionally associated with central nervous system function play critical roles in cancer. Dopamine receptor signaling is implicated in various cancers including glioblastoma (GBM) and it is a recognized therapeutic target, as evidenced by recent clinical trials with a selective dopamine receptor D2 (DRD2) inhibitor ONC201. Understanding the molecular mechanism(s) of the dopamine receptor signaling will be critical for development of potent therapeutic options. Using the human GBM patient-derived tumors treated with dopamine receptor agonists and antagonists, we identified the proteins that interact with DRD2. DRD2 signaling promotes glioblastoma (GBM) stem-like cells and GBM growth by activating MET. In contrast, pharmacological inhibition of DRD2 induces DRD2-TRAIL receptor interaction and subsequent cell death. Thus, our findings demonstrate a molecular circuitry of oncogenic DRD2 signaling in which MET and TRAIL receptors, critical factors for tumor cell survival and cell death, respectively, govern GBM survival and death. Finally, tumor-derived dopamine and expression of dopamine biosynthesis enzymes in a subset of GBM may guide patient stratification for DRD2 targeting therapy.
Collapse
Affiliation(s)
- Hye-Min Jeon
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Young Taek Oh
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Yong Jae Shin
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Nakho Chang
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Donggeun Kim
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Donghun Woo
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yoon Yeup
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Kyeung Min Joo
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Heejin Jo
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Heekyoung Yang
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Jin-Ku Lee
- Department of Biomedical Sciences, Department of Anatomy and Cell Biology, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Wonyoung Kang
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Jason Sa
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Won Jun Lee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James Hale
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Benjamin Purow
- Departments of Neurology, University of Virginia, Charlottesville, VA 22908, USA
| | - Myung Jin Park
- Divisions of Radiation Cancer Research, Research Center for Radio-Senescence, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Jong Bae Park
- Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Do-Hyun Nam
- Cancer Stem Cell Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Jeongwu Lee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
19
|
Farajizadeh F, Taghian F, Jalali Dehkordi K, Mirsafaei Rizi R. Swimming training and herbal nanoformulations as natural remedies to improve sensory-motor impairment in rat midbrain tumor models: system biology, behavioral test, and experimental validation. 3 Biotech 2023; 13:149. [PMID: 37131964 PMCID: PMC10148939 DOI: 10.1007/s13205-023-03574-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 04/19/2023] [Indexed: 05/04/2023] Open
Abstract
Motor impairment worsens health-related quality of life in patients with primary and metastatic midbrain tumors. Here, 56-male-Wistar rats were divided into eight groups: Normal group, Midbrain Tomur Model group, Model + Exe group, Model + Lipo, Model + Extract, Model + Lipo-Extract, Model + Extract-Exe, Model + Lipo-Extract + Exe. According to the aim, mid-brain tumor models were conducted by injections of the C6 glioma cell line (5 × 105 cell suspension) and stereotaxic techniques in the substantia nigra area. Furthermore, consumption of nanoformulation of herbals extract (100 mg/kg/day), crude extract (100 mg/kg/day), and swimming training (30 min, 3 days/week) as interventional protocols were performed for 6 weeks. In addition, we evaluated the effect of polyherbal nanoliposomes containing four plant extracts and swimming training on the GABArα1/TRKB/DRD2/DRD1a/TH network in the substantia nigra of the midbrain tumor rat model. Data emphasized that DRD2 might be a druggable protein with the network's highest significance cut-point effect that could modulate sensory-motor impairment. Furthermore, we found Quercetin, Ginsenosides, Curcumin, and Rutin, as bioactive compounds present in Ginseng, Matthiola incana, Turmeric, and Green-Tea extracts, could bind over the DRD2 protein with approved binding affinity scores. Based on our data, swimming training, and nanoliposome-enriched combined supplements could consider effective complementary medicine for motor impairment recovery induced by the midbrain tumor in the substantia nigra area. Hence, regular swimming training and natural medicines rich in polyphenolic bioactive components and antioxidative effects could modify and improve the dopamine receptors' function. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03574-3.
Collapse
Affiliation(s)
- Fariba Farajizadeh
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Farzaneh Taghian
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Khosro Jalali Dehkordi
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Rezvan Mirsafaei Rizi
- Department of Sports Injuries, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| |
Collapse
|
20
|
Sisakht AK, Malekan M, Ghobadinezhad F, Firouzabadi SNM, Jafari A, Mirazimi SMA, Abadi B, Shafabakhsh R, Mirzaei H. Cellular Conversations in Glioblastoma Progression, Diagnosis and Treatment. Cell Mol Neurobiol 2023; 43:585-603. [PMID: 35411434 DOI: 10.1007/s10571-022-01212-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 03/07/2022] [Indexed: 12/22/2022]
Abstract
Glioblastoma (GBM) is the most frequent malignancy among primary brain tumors in adults and one of the worst 5-year survival rates (< 7%) among all human cancers. Till now, treatments that target particular cell or intracellular metabolism have not improved patients' survival. GBM recruits healthy brain cells and subverts their processes to create a microenvironment that contributes to supporting tumor progression. This microenvironment encompasses a complex network in which malignant cells interact with each other and with normal and immune cells to promote tumor proliferation, angiogenesis, metastasis, immune suppression, and treatment resistance. Communication can be direct via cell-to-cell contact, mainly through adhesion molecules, tunneling nanotubes, gap junctions, or indirect by conventional paracrine signaling by cytokine, neurotransmitter, and extracellular vesicles. Understanding these communication routes could open up new avenues for the treatment of this lethal tumor. Hence, therapeutic approaches based on glioma cells` communication have recently drawn attention. This review summarizes recent findings on the crosstalk between glioblastoma cells and their tumor microenvironment, and the impact of this conversation on glioblastoma progression. We also discuss the mechanism of communication of glioma cells and their importance as therapeutic targets and diagnostic and prognostic biomarkers. Overall, understanding the biological mechanism of specific interactions in the tumor microenvironment may help in predicting patient prognosis and developing novel therapeutic strategies to target GBM.
Collapse
Affiliation(s)
- Ali Karimi Sisakht
- Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Malekan
- Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farbod Ghobadinezhad
- Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyedeh Negar Mousavi Firouzabadi
- Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.,Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Banafshe Abadi
- Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
21
|
Aporphine and isoquinoline derivatives block glioblastoma cell stemness and enhance temozolomide cytotoxicity. Sci Rep 2022; 12:21113. [PMID: 36477472 PMCID: PMC9729571 DOI: 10.1038/s41598-022-25534-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive and common primary malignant brain tumor with limited available therapeutic approaches. Despite improvements in therapeutic options for GBM patients, efforts to develop new successful strategies remain as major unmet medical needs. Based on the cytotoxic properties of aporphine compounds, we evaluated the biological effect of 12 compounds obtained through total synthesis of ( ±)-apomorphine hydrochloride (APO) against GBM cells. The compounds 2,2,2-trifluoro-1-(1-methylene-3,4-dihydroisoquinolin-2(1H)-yl)ethenone (A5) and ( ±)-1-(10,11-dimethoxy-6a,7-dihydro-4H-dibenzo[de,g]quinolin-6(5H)-yl)ethenone (C1) reduced the viability of GBM cells, with 50% inhibitory concentration ranging from 18 to 48 μM in patient-derived GBM cultures. Our data show that APO, A5 or C1 modulate the expression of DNA damage and apoptotic markers, impair 3D-gliomasphere growth and reduce the expression of stemness markers. Potential activity and protein targets of A5, C1 or APO were predicted in silico based on PASS and SEA software. Dopamine receptors (DRD1 and 5), CYP2B6, CYP2C9 and ABCB1, whose transcripts were differentially expressed in the GBM cells, were among the potential A5 or C1 target proteins. Docking analyses (HQSAR and 3D-QSAR) were performed to characterize possible interactions of ABCB1 and CYP2C9 with the compounds. Notably, A5 or C1 treatment, but not temozolomide (TMZ), reduced significantly the levels of extracellular ATP, suggesting ABCB1 negative regulation, which was correlated with stronger cytotoxicity induced by the combination of TMZ with A5 or C1 on GBM cells. Hence, our data reveal a potential therapeutic application of A5 and C1 as cytotoxic agents against GBM cells and predicted molecular networks that can be further exploited to characterize the pharmacological effects of these isoquinoline-containing substances.
Collapse
|
22
|
Zhang X, Zhao L, Zhang H, Zhang Y, Ju H, Wang X, Ren H, Zhu X, Dong Y. The immunosuppressive microenvironment and immunotherapy in human glioblastoma. Front Immunol 2022; 13:1003651. [PMID: 36466873 PMCID: PMC9712217 DOI: 10.3389/fimmu.2022.1003651] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/01/2022] [Indexed: 08/09/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant intracranial tumor in adults, characterized by extensive infiltrative growth, high vascularization, and resistance to multiple therapeutic approaches. Among the many factors affecting the therapeutic effect, the immunosuppressive GBM microenvironment that is created by cells and associated molecules via complex mechanisms plays a particularly important role in facilitating evasion of the tumor from the immune response. Accumulating evidence is also revealing a close association of the gut microbiota with the challenges in the treatment of GBM. The gut microbiota establishes a connection with the central nervous system through bidirectional signals of the gut-brain axis, thus affecting the occurrence and development of GBM. In this review, we discuss the key immunosuppressive components in the tumor microenvironment, along with the regulatory mechanism of the gut microbiota involved in immunity and metabolism in the GBM microenvironment. Lastly, we concentrate on the immunotherapeutic strategies currently under investigation, which hold promise to overcome the hurdles of the immunosuppressive tumor microenvironment and improve the therapeutic outcome for patients with GBM.
Collapse
Affiliation(s)
- Xuehua Zhang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Leilei Zhao
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - He Zhang
- Department of Immunology, Qiqihar Medical University, Qiqihar, China
| | - Yurui Zhang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Huanyu Ju
- Department of Immunology, Harbin Medical University, Harbin, China
| | - Xiaoyu Wang
- Department of Neurology, Hongda Hospital, Jinxiang, China
| | - Huan Ren
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiao Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, China
| | - Yucui Dong
- Department of Immunology, Binzhou Medical University, Yantai, China
| |
Collapse
|
23
|
Laskowska AK, Kleczkowska P. Anticancer efficacy of endo- and exogenous potent ligands acting at dopaminergic receptor-expressing cancer cells. Eur J Pharmacol 2022; 932:175230. [PMID: 36027983 DOI: 10.1016/j.ejphar.2022.175230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/27/2022]
Abstract
Cancer is one of the most common and dreaded diseases affecting the vastness of society. Unfortunately, still some people die especially when cancer is not diagnosed and thus caught early enough. On the other hand, using available chemo- or radiotherapy may result in serious side effects. Therefore, cancer-specific medications seem to be the most desired and safe therapy. Knowing that some cancers are characterized by overexpression of specific receptors on the cell surface, target-mediated drugs could serve as a unique and effective form of therapy. In line with this, recently dopaminergic receptors were presented important in cancer therapy as several dopaminergic ligands revealed their efficacy in tumor growth reduction as well as in apoptosis mediation. Unfortunately, the indication of whether DA receptor agonists or antagonists are the best choices in cancer treatment is quite difficult, since both of them may exert either pro- or anticancer effects. In this review, we analyze the therapeutic efficacy of compounds, both of exogenous and endogenous origin, targeting dopaminergic receptor-expressing cancers.
Collapse
Affiliation(s)
- Anna K Laskowska
- Centre for Preclinical Research and Technology (CePT), Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1B Str., 02-097, Warsaw, Poland
| | - Patrycja Kleczkowska
- Maria Sklodowska-Curie Medical Academy in Warsaw, Solidarnosci 12 Str., 03-411, Warsaw, Poland; Military Institute of Hygiene and Epidemiology, Kozielska 4 Str., 01-163, Warsaw, Poland.
| |
Collapse
|
24
|
Neurotransmitters: Potential Targets in Glioblastoma. Cancers (Basel) 2022; 14:cancers14163970. [PMID: 36010960 PMCID: PMC9406056 DOI: 10.3390/cancers14163970] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/01/2022] [Accepted: 08/12/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Aiming to discover potential treatments for GBM, this review connects emerging research on the roles of neurotransmitters in the normal neural and the GBM microenvironments and sheds light on the prospects of their application in the neuropharmacology of GBM. Conventional therapy is blamed for its poor effect, especially in inhibiting tumor recurrence and invasion. Facing this dilemma, we focus on neurotransmitters that modulate GBM initiation, progression and invasion, hoping to provide novel therapy targeting GBM. By analyzing research concerning GBM therapy systematically and scientifically, we discover increasing insights into the regulatory effects of neurotransmitters, some of which have already shown great potential in research in vivo or in vitro. After that, we further summarize the potential drugs in correlation with previously published research. In summary, it is worth expecting that targeting neurotransmitters could be a promising novel pharmacological approach for GBM treatment. Abstract For decades, glioblastoma multiforme (GBM), a type of the most lethal brain tumor, has remained a formidable challenge in terms of its treatment. Recently, many novel discoveries have underlined the regulatory roles of neurotransmitters in the microenvironment both physiologically and pathologically. By targeting the receptors synaptically or non-synaptically, neurotransmitters activate multiple signaling pathways. Significantly, many ligands acting on neurotransmitter receptors have shown great potential for inhibiting GBM growth and development, requiring further research. Here, we provide an overview of the most novel advances concerning the role of neurotransmitters in the normal neural and the GBM microenvironments, and discuss potential targeted drugs used for GBM treatment.
Collapse
|
25
|
Vachalova V, Karahoda R, Ottaviani M, Anandam KY, Abad C, Albrecht C, Staud F. Functional reorganization of monoamine transport systems during villous trophoblast differentiation: evidence of distinct differences between primary human trophoblasts and BeWo cells. Reprod Biol Endocrinol 2022; 20:112. [PMID: 35927731 PMCID: PMC9351077 DOI: 10.1186/s12958-022-00981-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Three primary monoamines-serotonin, norepinephrine, and dopamine-play major roles in the placenta-fetal brain axis. Analogously to the brain, the placenta has transport mechanisms that actively take up these monoamines into trophoblast cells. These transporters are known to play important roles in the differentiated syncytiotrophoblast layer, but their status and activities in the undifferentiated, progenitor cytotrophoblast cells are not well understood. Thus, we have explored the cellular handling and regulation of monoamine transporters during the phenotypic transitioning of cytotrophoblasts along the villous pathway. METHODS Experiments were conducted with two cellular models of syncytium development: primary trophoblast cells isolated from the human term placenta (PHT), and the choriocarcinoma-derived BeWo cell line. The gene and protein expression of membrane transporters for serotonin (SERT), norepinephrine (NET), dopamine (DAT), and organic cation transporter 3 (OCT3) was determined by quantitative PCR and Western blot analysis, respectively. Subsequently, the effect of trophoblast differentiation on transporter activity was analyzed by monoamine uptake into cells. RESULTS We present multiple lines of evidence of changes in the transcriptional and functional regulation of monoamine transporters associated with trophoblast differentiation. These include enhancement of SERT and DAT gene and protein expression in BeWo cells. On the other hand, in PHT cells we report negative modulation of SERT, NET, and OCT3 protein expression. We show that OCT3 is the dominant monoamine transporter in PHT cells, and its main functional impact is on serotonin uptake, while passive transport strongly contributes to norepinephrine and dopamine uptake. Further, we show that a wide range of selective serotonin reuptake inhibitors affect serotonin cellular accumulation, at pharmacologically relevant drug concentrations, via their action on both OCT3 and SERT. Finally, we demonstrate that BeWo cells do not well reflect the molecular mechanisms and properties of healthy human trophoblast cells. CONCLUSIONS Collectively, our findings provide insights into the regulation of monoamine transport during trophoblast differentiation and present important considerations regarding appropriate in vitro models for studying monoamine regulation in the placenta.
Collapse
Affiliation(s)
- Veronika Vachalova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Martina Ottaviani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Kasin Yadunandam Anandam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
- Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
26
|
Uthamacumaran A. Dissecting cell fate dynamics in pediatric glioblastoma through the lens of complex systems and cellular cybernetics. BIOLOGICAL CYBERNETICS 2022; 116:407-445. [PMID: 35678918 DOI: 10.1007/s00422-022-00935-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Cancers are complex dynamic ecosystems. Reductionist approaches to science are inadequate in characterizing their self-organized patterns and collective emergent behaviors. Since current approaches to single-cell analysis in cancer systems rely primarily on single time-point multiomics, many of the temporal features and causal adaptive behaviors in cancer dynamics are vastly ignored. As such, tools and concepts from the interdisciplinary paradigm of complex systems theory are introduced herein to decode the cellular cybernetics of cancer differentiation dynamics and behavioral patterns. An intuition for the attractors and complex networks underlying cancer processes such as cell fate decision-making, multiscale pattern formation systems, and epigenetic state-transitions is developed. The applications of complex systems physics in paving targeted therapies and causal pattern discovery in precision oncology are discussed. Pediatric high-grade gliomas are discussed as a model-system to demonstrate that cancers are complex adaptive systems, in which the emergence and selection of heterogeneous cellular states and phenotypic plasticity are driven by complex multiscale network dynamics. In specific, pediatric glioblastoma (GBM) is used as a proof-of-concept model to illustrate the applications of the complex systems framework in understanding GBM cell fate decisions and decoding their adaptive cellular dynamics. The scope of these tools in forecasting cancer cell fate dynamics in the emerging field of computational oncology and patient-centered systems medicine is highlighted.
Collapse
|
27
|
Persico M, Abbruzzese C, Matteoni S, Matarrese P, Campana AM, Villani V, Pace A, Paggi MG. Tackling the Behavior of Cancer Cells: Molecular Bases for Repurposing Antipsychotic Drugs in the Treatment of Glioblastoma. Cells 2022; 11:263. [PMID: 35053377 PMCID: PMC8773942 DOI: 10.3390/cells11020263] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma (GBM) is associated with a very dismal prognosis, and current therapeutic options still retain an overall unsatisfactorily efficacy in clinical practice. Therefore, novel therapeutic approaches and effective medications are highly needed. Since the development of new drugs is an extremely long, complex and expensive process, researchers and clinicians are increasingly considering drug repositioning/repurposing as a valid alternative to the standard research process. Drug repurposing is also under active investigation in GBM therapy, since a wide range of noncancer and cancer therapeutics have been proposed or investigated in clinical trials. Among these, a remarkable role is played by the antipsychotic drugs, thanks to some still partially unexplored, interesting features of these agents. Indeed, antipsychotic drugs have been described to interfere at variable incisiveness with most hallmarks of cancer. In this review, we analyze the effects of antipsychotics in oncology and how these drugs can interfere with the hallmarks of cancer in GBM. Overall, according to available evidence, mostly at the preclinical level, it is possible to speculate that repurposing of antipsychotics in GBM therapy might contribute to providing potentially effective and inexpensive therapies for patients with this disease.
Collapse
Affiliation(s)
- Michele Persico
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| | - Claudia Abbruzzese
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| | - Silvia Matteoni
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| | - Paola Matarrese
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, 00162 Rome, Italy;
| | - Anna Maria Campana
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA;
| | - Veronica Villani
- Neuro-Oncology, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.V.); (A.P.)
| | - Andrea Pace
- Neuro-Oncology, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.V.); (A.P.)
| | - Marco G. Paggi
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| |
Collapse
|
28
|
Zhu X, Pan S, Li R, Chen Z, Xie X, Han D, Lv S, Huang Y. Novel Biomarker Genes for Prognosis of Survival and Treatment of Glioma. Front Oncol 2022; 11:667884. [PMID: 34976783 PMCID: PMC8714878 DOI: 10.3389/fonc.2021.667884] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive malignant primary central nervous system tumor. Although surgery, radiotherapy, and chemotherapy treatments are available, the 5-year survival rate of GBM is only 5.8%. Therefore, it is imperative to find novel biomarker for the prognosis and treatment of GBM. In this study, a total of 141 differentially expressed genes (DEGs) in GBM were identified by analyzing the GSE12657, GSE90886, and GSE90598 datasets. After reducing the data dimensionality, Kaplan-Meier survival analysis indicated that expression of PTPRN and RIM-BP2 were downregulated in GBM tissues when compared with that of normal tissues and that the expression of these genes was a good prognostic biomarker for GBM (p<0.05). Then, the GSE46531 dataset and the Genomics of Drug Sensitivity in Cancer (GDSC) database were used to examine the relationship between sensitivity radiotherapy (RT) and chemotherapy for GBM and expression of PTPRN and RIM-BP2. The expression of PTPRN was significantly high in RT-resistant patients (p<0.05) but it was not related to temozolomide (TMZ) resistance. The expression level of RIM-BP2 was not associated with RT or TMZ treatment. Among the chemotherapeutic drugs, cisplatin and erlotinib had a significantly good treatment effect for glioma with expression of PTPRN or RIM-BP2 and in lower-grade glioma (LGG) with IDH mutation. (p < 0.05). The tumor mutational burden (TMB) score in the low PTPRN expression group was significantly higher than that in the high PTPRN expression group (p=0.013), with a large degree of tumor immune cell infiltration. In conclusion, these findings contributed to the discovery process of potential biomarkers and therapeutic targets for glioma patients.
Collapse
Affiliation(s)
- Xiaopeng Zhu
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Sian Pan
- Department of Rehabilitation Medicine, Zhuzhou Central Hospital, Zhuzhou, China
| | - Rui Li
- Department of Operating Theatre, Zhuzhou Central Hospital, Zhuzhou, China
| | - Zebo Chen
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Xingyun Xie
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Deqing Han
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Shengqing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yongkai Huang
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, China
| |
Collapse
|
29
|
Björkblom B, Wibom C, Eriksson M, Bergenheim AT, Sjöberg RL, Jonsson P, Brännström T, Antti H, Sandström M, Melin B. OUP accepted manuscript. Neuro Oncol 2022; 24:1454-1468. [PMID: 35157758 PMCID: PMC9435506 DOI: 10.1093/neuonc/noac042] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Benny Björkblom
- Corresponding Author: Dr. Benny Björkblom, PhD, Department of Chemistry, Umeå University, Linnaeus väg 10, SE-901 87 Umeå, Sweden ()
| | - Carl Wibom
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Maria Eriksson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - A Tommy Bergenheim
- Department of Clinical Science, Neuroscience, Umeå University, Umeå, Sweden
| | - Rickard L Sjöberg
- Department of Clinical Science, Neuroscience, Umeå University, Umeå, Sweden
| | - Pär Jonsson
- Department of Chemistry, Umeå University, Umeå, Sweden
| | | | - Henrik Antti
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Maria Sandström
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Beatrice Melin
- Corresponding Author: Professor Beatrice Melin, MD, PhD, Department of Radiation Sciences, Oncology, Umeå University, SE-901 87 Umeå, Sweden ()
| |
Collapse
|
30
|
Guo X, Piao H. Research Progress of circRNAs in Glioblastoma. Front Cell Dev Biol 2021; 9:791892. [PMID: 34881248 PMCID: PMC8645988 DOI: 10.3389/fcell.2021.791892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/02/2021] [Indexed: 01/10/2023] Open
Abstract
Circular RNAs (circRNAs) are a class of single-stranded covalently closed non-coding RNAs without a 5' cap structure or 3' terminal poly (A) tail, which are expressed in a variety of tissues and cells with conserved, stable and specific characteristics. Glioblastoma (GBM) is the most aggressive and lethal tumor in the central nervous system, characterized by high recurrence and mortality rates. The specific expression of circRNAs in GBM has demonstrated their potential to become new biomarkers for the development of GBM. The specific expression of circRNAs in GBM has shown their potential as new biomarkers for GBM cell proliferation, apoptosis, migration and invasion, which provides new ideas for GBM treatment. In this paper, we will review the biological properties and functions of circRNAs and their biological roles and clinical applications in GBM.
Collapse
Affiliation(s)
- Xu Guo
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Haozhe Piao
- Department of Neurosurgery, Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| |
Collapse
|
31
|
Kirby AJ, Finnerty GT. New strategies for managing adult gliomas. J Neurol 2021; 268:3666-3674. [PMID: 32542524 PMCID: PMC8463358 DOI: 10.1007/s00415-020-09884-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 12/19/2022]
Abstract
Gliomas are hard to treat. Their prognosis has improved little over the past few decades. Fundamental therapeutic challenges such as treatment resistance, malignant progression, and tumour recurrence persist. New strategies are needed to advance the management and treatment of gliomas. Here, we focus on where those new strategies could emerge. We consider how recent advances in our understanding of the biology of adult gliomas are informing new approaches to their treatment.
Collapse
Affiliation(s)
- Alastair J Kirby
- Department of Basic and Clinical Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Gerald T Finnerty
- Department of Basic and Clinical Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK.
- Department of Neurology, King's College Hospital NHS Foundation Trust, Denmark Hill, London, SE5 9RS, UK.
| |
Collapse
|
32
|
Sarrouilhe D, Defamie N, Mesnil M. Is the Exposome Involved in Brain Disorders through the Serotoninergic System? Biomedicines 2021; 9:1351. [PMID: 34680468 PMCID: PMC8533279 DOI: 10.3390/biomedicines9101351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/17/2021] [Accepted: 09/23/2021] [Indexed: 11/24/2022] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a biogenic monoamine acting as a neurotransmitter in the central nervous system (CNS), local mediator in the gut, and vasoactive agent in the blood. It has been linked to a variety of CNS functions and is implicated in many CNS and psychiatric disorders. The high comorbidity between some neuropathies can be partially understood by the fact that these diseases share a common etiology involving the serotoninergic system. In addition to its well-known functions, serotonin has been shown to be a mitogenic factor for a wide range of normal and tumor cells, including glioma cells, in vitro. The developing CNS of fetus and newborn is particularly susceptible to the deleterious effects of neurotoxic substances in our environment, and perinatal exposure could result in the later development of diseases, a hypothesis known as the developmental origin of health and disease. Some of these substances affect the serotoninergic system and could therefore be the source of a silent pandemic of neurodevelopmental toxicity. This review presents the available data that are contributing to the appreciation of the effects of the exposome on the serotoninergic system and their potential link with brain pathologies (neurodevelopmental, neurodegenerative, neurobehavioral disorders, and glioblastoma).
Collapse
Affiliation(s)
- Denis Sarrouilhe
- Laboratoire de Physiologie Humaine, Faculté de Médecine et Pharmacie, 6 Rue de la Milétrie, Bât D1, TSA 51115, CEDEX 09, 86073 Poitiers, France
| | - Norah Defamie
- Laboratoire STIM, ERL7003 CNRS-Université de Poitiers, 1 Rue G. Bonnet–TSA 51106, CEDEX 09, 86073 Poitiers, France; (N.D.); (M.M.)
| | - Marc Mesnil
- Laboratoire STIM, ERL7003 CNRS-Université de Poitiers, 1 Rue G. Bonnet–TSA 51106, CEDEX 09, 86073 Poitiers, France; (N.D.); (M.M.)
| |
Collapse
|
33
|
Liu H, Sun Y, Zhang Q, Jin W, Gordon RE, Zhang Y, Wang J, Sun C, Wang ZJ, Qi X, Zhang J, Huang B, Gui Q, Yuan H, Chen L, Ma X, Fang C, Liu YQ, Yu X, Feng S. Pro-inflammatory and proliferative microglia drive progression of glioblastoma. Cell Rep 2021; 36:109718. [PMID: 34525361 DOI: 10.1016/j.celrep.2021.109718] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 04/01/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022] Open
Abstract
Scant understanding of the glioblastoma microenvironment and molecular bases hampers development of efficient treatment strategies. Analyses of gene signatures of human gliomas demonstrate that the SETD2 mutation is correlated with poor prognosis of IDH1/2 wild-type (IDH-WT) adult glioblastoma patients. To better understand the crosstalk between SETD2 mutant (SETD2-mut) glioblastoma cells and the tumor microenvironment, we leverage single-cell transcriptomics to comprehensively map cellular populations in glioblastoma. In this study, we identify a specific subtype of high-grade glioma-associated microglia (HGG-AM). Further analysis shows that transforming growth factor (TGF)-β1 derived from SETD2-mut/IDH-WT tumor cells activates HGG-AM, exhibiting pro-inflammation and proliferation signatures. Particularly, HGG-AM secretes interleukin (IL)-1β via the apolipoprotein E (ApoE)-mediated NLRP1 inflammasome, thereby promoting tumor progression. HGG-AM present extensive proliferation and infiltration to supplement the activated microglia pool. Notably, TGF-β1/TβRI depletion dramatically reduces HGG-AM density and suppresses tumor growth. Altogether, our studies identify a specific microglia subpopulation and establish the cellular basis of interactions between HGG-AM and glioblastoma cells.
Collapse
Affiliation(s)
- Hailong Liu
- Department of Neurosurgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China; Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing 10070, P.R. China
| | - Youliang Sun
- School of Basic Medical Science, Capital Medical University, Beijing 100069, P.R. China
| | - Qian Zhang
- Medical Laboratory Center, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, P.R. China; Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518000, P.R. China
| | - Wei Jin
- Department of Pathology, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | | | - Yanyang Zhang
- Department of Neurosurgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Jian Wang
- Department of Neurosurgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Caihong Sun
- Department of Neurosurgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Zeyuan John Wang
- School of Pharmaceutical Sciences, Temple University, Philadelphia, PA 19140, USA
| | - Xueling Qi
- Department of Neuro-Pathology, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P. R. China
| | - Junping Zhang
- Department of Neuro-Oncology, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| | - Boyuan Huang
- Department of Neurosurgery, Beijing Electric Power Hospital, Beijing 100073, P.R. China
| | - Qiuping Gui
- Department of Pathology, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Hongyu Yuan
- State Key Laboratory of Molecular Oncology, Chinese Academy of Medical Science Cancer Hospital/National Cancer Center, Beijing 100021, P.R. China
| | - Ling Chen
- Department of Neurosurgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Xiaodong Ma
- Department of Neurosurgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Chuan Fang
- Department of Neurosurgery, The Affiliated Hospital of Hebei University, Baoding 122311, P.R. China
| | - Yong-Qiang Liu
- Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China.
| | - Xinguang Yu
- Department of Neurosurgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China.
| | - Shiyu Feng
- Department of Neurosurgery, The First Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China.
| |
Collapse
|
34
|
Influence of scalp block on oncological outcomes of high-grade glioma in adult patients with and without isocitrate dehydrogenase-1 mutation. Sci Rep 2021; 11:16489. [PMID: 34389754 PMCID: PMC8363618 DOI: 10.1038/s41598-021-95851-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/27/2021] [Indexed: 11/08/2022] Open
Abstract
High-grade gliomas are notorious for a high recurrence rate even after curative resection surgery. Studies regarding the influence of scalp block on high-grade gliomas have been inconclusive, possibly because the condition's most important genetic mutation profile, namely the isocitrate dehydrogenase 1 (IDH1) mutation, had not been analyzed. Therefore, we conducted a single-center study including patients with high-grade glioma who underwent tumor resection between January 2014 and December 2019. Kaplan-Meier survival analysis revealed that scalp block was associated with longer progression-free survival (PFS; 15.17 vs. 10.77 months, p = 0.0018), as was the IDH1 mutation (37.37 vs. 10.90 months, p = 0.0149). Multivariate Cox regression analysis revealed that scalp block (hazard ratio: 0.436, 95% confidence interval: 0.236-0.807, p = 0.0082), gross total resection (hazard ratio: 0.405, 95% confidence interval: 0.227-0.721, p = 0.0021), and IDH1 mutation (hazard ratio: 0.304, 95% confidence interval: 0.118-0.784, p = 0.0138) were associated with better PFS. Our results demonstrate that application of scalp block, regardless of IDH1 profile, is an independent factor associated with longer PFS for patients with high-grade glioma.
Collapse
|
35
|
Perrone MG, Ruggiero A, Centonze A, Carrieri A, Ferorelli S, Scilimati A. Diffuse Intrinsic Pontine Glioma (DIPG): Breakthrough and Clinical Perspective. Curr Med Chem 2021; 28:3287-3317. [PMID: 32767913 DOI: 10.2174/0929867327666200806110206] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/16/2020] [Accepted: 07/23/2020] [Indexed: 11/22/2022]
Abstract
Diffuse intrinsic pontine glioma (DIPG) mainly affects children with a median age of 6-7 years old. It accounts for 10% of all pediatric tumors. Unfortunately, DIPG has a poor prognosis, and the median survival is generally less than 16-24 months independently from the treatment received. Up to now, children with DIPG are treated with focal radiotherapy alone or in combination with antitumor agents. In the last decade, ONC201 known as dopamine receptor antagonist was uncovered, by a high throughput screening of public libraries of compounds, to be endowed with cytotoxic activity against several cancer cell lines. Efforts were made to identify the real ONC201 target, responsible for its antiproliferative effect. The hypothesized targets were the Tumor necrosis factor-Related Apoptosis-Inducing Ligand stimulation (TRAIL), two oncogenic kinases (ERK/AKT system) that target the same tumor-suppressor gene (FOXO3a), dopamine receptors (DRD2 and DRD3 subtypes) and finally the mitochondrial Caseynolitic Protease P (ClpP). ONC201 structure-activity relationship is extensively discussed in this review, together with other two classes of compounds, namely ADEPs and D9, already known for their antibiotic activity but noteworthy to be discussed and studied as potential "leads" for the development of new drugs to be used in the treatment of DIPG. In this review, a detailed and critical description of ONC201, ADEPs, and D9 pro-apoptotic activity is made, with particular attention to the specific interactions established with its targets that also are intimately described. Pubmed published patents and clinical trial reports of the last ten years were used as the bibliographic source.
Collapse
Affiliation(s)
- Maria Grazia Perrone
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Antonella Centonze
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | - Antonio Carrieri
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | - Savina Ferorelli
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| | - Antonio Scilimati
- Department of Pharmacy and Pharmaceutical Sciences, University of Bari, Via E. Orabona 4, 70125 Bari, Italy
| |
Collapse
|
36
|
D’Alessandro G, Lauro C, Quaglio D, Ghirga F, Botta B, Trettel F, Limatola C. Neuro-Signals from Gut Microbiota: Perspectives for Brain Glioma. Cancers (Basel) 2021; 13:2810. [PMID: 34199968 PMCID: PMC8200200 DOI: 10.3390/cancers13112810] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/25/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive form of glioma tumor in adult brain. Among the numerous factors responsible for GBM cell proliferation and invasion, neurotransmitters such as dopamine, serotonin and glutamate can play key roles. Studies performed in mice housed in germ-free (GF) conditions demonstrated the relevance of the gut-brain axis in a number of physiological and pathological conditions. The gut-brain communication is made possible by vagal/nervous and blood/lymphatic routes and pave the way for reciprocal modulation of functions. The gut microbiota produces and consumes a wide range of molecules, including neurotransmitters (dopamine, norepinephrine, serotonin, gamma-aminobutyric acid [GABA], and glutamate) that reach their cellular targets through the bloodstream. Growing evidence in animals suggests that modulation of these neurotransmitters by the microbiota impacts host neurophysiology and behavior, and affects neural cell progenitors and glial cells, along with having effects on tumor cell growth. In this review we propose a new perspective connecting neurotransmitter modulation by gut microbiota to glioma progression.
Collapse
Affiliation(s)
- Giuseppina D’Alessandro
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy; (G.D.); (C.L.); (F.T.)
- IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy; (G.D.); (C.L.); (F.T.)
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.Q.); (F.G.); (B.B.)
| | - Francesca Ghirga
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.Q.); (F.G.); (B.B.)
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University, P.le Aldo Moro 5, 00185 Rome, Italy; (D.Q.); (F.G.); (B.B.)
| | - Flavia Trettel
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy; (G.D.); (C.L.); (F.T.)
| | - Cristina Limatola
- IRCCS Neuromed, 86077 Pozzilli, IS, Italy
- Department of Physiology and Pharmacology, Sapienza University, Laboratory Affiliated to Istituto Pasteur Italia, 00185 Rome, Italy
| |
Collapse
|
37
|
Platten M, Friedrich M, Wainwright DA, Panitz V, Opitz CA. Tryptophan metabolism in brain tumors - IDO and beyond. Curr Opin Immunol 2021; 70:57-66. [PMID: 33813026 DOI: 10.1016/j.coi.2021.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/27/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022]
Abstract
Metabolism of the essential amino acid tryptophan is a key metabolic pathway that restricts antitumor immunity and is a drug development target for cancer immunotherapy. Tryptophan metabolism is active in brain tumors including gliomas and promotes a malignant phenotype and contributes to the immunosuppressive tumor microenvironment. In recent years, improved understanding of the regulation and downstream function of tryptophan metabolism has been significantly expanded beyond the initial in vitro observation that the enzyme indoleamine-2,3-dioxygenase 1 (IDO1) promotes the depletion of intracellular tryptophan. Here, we revisit the specific roles of tryptophan metabolites in regulating brain functioning and neuronal integrity as well as in the context of brain tumors. This review summarizes recent developments in identifying key regulators, as well as the cellular and molecular effects of tryptophan metabolism with a particular focus on potential therapeutic targets in glioma.
Collapse
Affiliation(s)
- Michael Platten
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Heidelberg, Germany; DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Mirco Friedrich
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Heidelberg, Germany; DKTK CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Derek A Wainwright
- Departments of Neurological Surgery, Medicine - Division of Hematology/Oncology, and Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Verena Panitz
- DKTK Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Christiane A Opitz
- DKTK Brain Cancer Metabolism Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
38
|
Arrillaga-Romany I, Odia Y, Prabhu VV, Tarapore RS, Merdinger K, Stogniew M, Oster W, Allen JE, Mehta M, Batchelor TT, Wen PY. Biological activity of weekly ONC201 in adult recurrent glioblastoma patients. Neuro Oncol 2021; 22:94-102. [PMID: 31702782 DOI: 10.1093/neuonc/noz164] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND ONC201 is a dopamine receptor D2 (DRD2) antagonist that penetrates the blood-brain barrier. ONC201 efficacy has been shown in glioblastoma animal models and is inversely correlated with dopamine receptor DRD5 expression. ONC201 is well tolerated in adult recurrent glioblastoma patients with dosing every 3 weeks and has achieved an objective radiographic response in a patient harboring the H3 K27M mutation. METHODS In a window-of-opportunity arm, 6 adult subjects initiated ONC201 prior to re-resection of recurrent glioblastoma with intratumoral concentrations as the primary endpoint. An additional 20 adults with recurrent glioblastoma received single agent weekly oral ONC201 at 625 mg, with progression-free survival at 6 months (PFS6) by Response Assessment in Neuro-Oncology (RANO) criteria as the primary endpoint. RESULTS The window-of-opportunity arm achieved its primary endpoint with intratumoral ONC201 concentrations at ~24 hours following the second weekly dose ranging from 600 nM to 9.3 µM. Intratumoral pharmacodynamics assessed by activating transcriptional factor 4, death receptor 5, and apoptosis induction relative to archival samples were observed with the strongest intensity and uniformity among patients with low DRD5 tumor expression. The primary endpoint of PFS6 by RANO was not achieved at 5% in this molecularly unselected cohort; however, 1 of 3 patients enrolled with the H3 K27M mutation had a complete regression of enhancing multifocal lesions that remained durable for >1.5 years. No treatment modifications or discontinuations due to toxicity were observed, including in those who underwent re-resection. CONCLUSIONS Weekly ONC201 is well tolerated, and meaningful intratumoral concentrations were achieved. ONC201 may be biologically active in a subset of adult patients with recurrent glioblastoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tracy T Batchelor
- Brigham and Women's Hospital, Boston, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | | |
Collapse
|
39
|
Tan JY, Wijesinghe IVS, Alfarizal Kamarudin MN, Parhar I. Paediatric Gliomas: BRAF and Histone H3 as Biomarkers, Therapy and Perspective of Liquid Biopsies. Cancers (Basel) 2021; 13:cancers13040607. [PMID: 33557011 PMCID: PMC7913734 DOI: 10.3390/cancers13040607] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Gliomas are major causes of worldwide cancer-associated deaths in children. Generally, paediatric gliomas can be classified into low-grade and high-grade gliomas. They differ significantly from adult gliomas in terms of prevalence, molecular alterations, molecular mechanisms and predominant histological types. The aims of this review article are: (i) to discuss the current updates of biomarkers in paediatric low-grade and high-grade gliomas including their diagnostic and prognostic values, and (ii) to discuss potential targeted therapies in treating paediatric low-grade and high-grade gliomas. Our findings revealed that liquid biopsy is less invasive than tissue biopsy in obtaining the samples for biomarker detections in children. In addition, future clinical trials should consider blood-brain barrier (BBB) penetration of therapeutic drugs in paediatric population. Abstract Paediatric gliomas categorised as low- or high-grade vary markedly from their adult counterparts, and denoted as the second most prevalent childhood cancers after leukaemia. As compared to adult gliomas, the studies of diagnostic and prognostic biomarkers, as well as the development of therapy in paediatric gliomas, are still in their infancy. A body of evidence demonstrates that B-Raf Proto-Oncogene or V-Raf Murine Sarcoma Viral Oncogene Homolog B (BRAF) and histone H3 mutations are valuable biomarkers for paediatric low-grade gliomas (pLGGs) and high-grade gliomas (pHGGs). Various diagnostic methods involving fluorescence in situ hybridisation, whole-genomic sequencing, PCR, next-generation sequencing and NanoString are currently used for detecting BRAF and histone H3 mutations. Additionally, liquid biopsies are gaining popularity as an alternative to tumour materials in detecting these biomarkers, but still, they cannot fully replace solid biopsies due to several limitations. Although histone H3 mutations are reliable prognosis biomarkers in pHGGs, children with these mutations have a dismal prognosis. Conversely, the role of BRAF alterations as prognostic biomarkers in pLGGs is still in doubt due to contradictory findings. The BRAF V600E mutation is seen in the majority of pLGGs (as seen in pleomorphic xanthoastrocytoma and gangliomas). By contrast, the H3K27M mutation is found in the majority of paediatric diffuse intrinsic pontine glioma and other midline gliomas in pHGGs. pLGG patients with a BRAF V600E mutation often have a lower progression-free survival rate in comparison to wild-type pLGGs when treated with conventional therapies. BRAF inhibitors (Dabrafenib and Vemurafenib), however, show higher overall survival and tumour response in BRAF V600E mutated pLGGs than conventional therapies in some studies. To date, targeted therapy and precision medicine are promising avenues for paediatric gliomas with BRAF V600E and diffuse intrinsic pontine glioma with the H3K27M mutations. Given these shortcomings in the current treatments of paediatric gliomas, there is a dire need for novel therapies that yield a better therapeutic response. The present review discusses the diagnostic tools and the perspective of liquid biopsies in the detection of BRAF V600E and H3K27M mutations. An in-depth understanding of these biomarkers and the therapeutics associated with the respective challenges will bridge the gap between paediatric glioma patients and the development of effective therapies.
Collapse
Affiliation(s)
| | | | | | - Ishwar Parhar
- Correspondence: ; Tel.: +603-5514-6304; Fax: +603-5515-6341
| |
Collapse
|
40
|
Rosas-Cruz A, Salinas-Jazmín N, Velázquez MAV. Dopamine Receptors in Cancer: Are They Valid Therapeutic Targets? Technol Cancer Res Treat 2021; 20:15330338211027913. [PMID: 34212819 PMCID: PMC8255587 DOI: 10.1177/15330338211027913] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/27/2021] [Accepted: 05/18/2021] [Indexed: 12/24/2022] Open
Abstract
The dopamine receptors (DRs) family includes 5 members with differences in signal transduction and ligand affinity. Abnormal DRs expression has been correlated multiple tumors with their clinical outcome. Thus, it has been proposed that DRs-targeting drugs-developed for other diseases as schizophrenia or Parkinson's disease-could be helpful in managing neoplastic diseases. In this review, we discuss the role of DRs and the effects of DRs-targeting in tumor progression and cancer cell biology using multiple high-prevalence neoplasms as examples. The evidence shows that DRs are valid therapeutic targets for certain receptor/disease combinations, but the data are inconclusive or contradictory for others. In either case, further studies are required to define the precise role of DRs in tumor progression and propose better therapeutic strategies for their targeting.
Collapse
Affiliation(s)
- Arely Rosas-Cruz
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), México
- Posgrado en Ciencias Bioquímicas, UNAM, México
| | - Nohemí Salinas-Jazmín
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), México
| | - Marco A. Velasco- Velázquez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), México
- Unidad Periférica de Investigación en Biomedicina Traslacional, Centro Médico Nacional 20 de noviembre ISSSTE / Facultad de Medicina, UNAM, México
| |
Collapse
|
41
|
Kraboth Z, Galik B, Tompa M, Kajtar B, Urban P, Gyenesei A, Miseta A, Kalman B. DNA CpG methylation in sequential glioblastoma specimens. J Cancer Res Clin Oncol 2020; 146:2885-2896. [PMID: 32779022 PMCID: PMC7519911 DOI: 10.1007/s00432-020-03349-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/04/2020] [Indexed: 01/21/2023]
Abstract
PURPOSE Glioblastoma is the most aggressive form of brain tumors. A better understanding of the molecular mechanisms leading to its evolution is essential for the development of treatments more effective than the available modalities. Here, we aim to identify molecular drivers of glioblastoma development and recurrence by analyzing DNA CpG methylation patterns in sequential samples. METHODS DNA was isolated from 22 pairs of primary and recurrent formalin-fixed, paraffin-embedded glioblastoma specimens, and subjected to reduced representation bisulfite sequencing. Bioinformatic analyses were conducted to identify differentially methylated sites and pathways, and biostatistics was used to test correlations among clinical and pathological parameters. RESULTS Differentially methylated pathways likely involved in primary tumor development included those of neuronal differentiation, myelination, metabolic processes, synapse organization and endothelial cell proliferation, while pathways differentially active during glioblastoma recurrence involved those associated with cell processes and differentiation, immune response, Wnt regulation and catecholamine secretion and transport. CONCLUSION DNA CpG methylation analyses in sequential clinical specimens revealed hypomethylation in certain pathways such as neuronal tissue development and angiogenesis likely involved in early tumor development and growth, while suggested altered regulation in catecholamine secretion and transport, Wnt expression and immune response contributing to glioblastoma recurrence. These pathways merit further investigations and may represent novel therapeutic targets.
Collapse
Affiliation(s)
- Zoltan Kraboth
- Institute of Laboratory Medicine, School of Medicine, University of Pecs, Pecs, Hungary
- Szentagothai Research Center, University of Pecs, 20. Ifjusag street, Pecs, 7624, Hungary
| | - Bence Galik
- Szentagothai Research Center, University of Pecs, 20. Ifjusag street, Pecs, 7624, Hungary
- Department of Clinical Molecular Biology, Medical University of Bialystok, Białystok, Poland
| | - Marton Tompa
- Institute of Laboratory Medicine, School of Medicine, University of Pecs, Pecs, Hungary
- Szentagothai Research Center, University of Pecs, 20. Ifjusag street, Pecs, 7624, Hungary
| | - Bela Kajtar
- Institute of Pathology, School of Medicine, University of Pecs, Pecs, Hungary
| | - Peter Urban
- Szentagothai Research Center, University of Pecs, 20. Ifjusag street, Pecs, 7624, Hungary
| | - Attila Gyenesei
- Szentagothai Research Center, University of Pecs, 20. Ifjusag street, Pecs, 7624, Hungary
- Department of Clinical Molecular Biology, Medical University of Bialystok, Białystok, Poland
| | - Attila Miseta
- Institute of Laboratory Medicine, School of Medicine, University of Pecs, Pecs, Hungary
| | - Bernadette Kalman
- Institute of Laboratory Medicine, School of Medicine, University of Pecs, Pecs, Hungary.
- Szentagothai Research Center, University of Pecs, 20. Ifjusag street, Pecs, 7624, Hungary.
| |
Collapse
|
42
|
Dono A, Patrizz A, McCormack RM, Putluri N, Ganesh BP, Kaur B, McCullough LD, Ballester LY, Esquenazi Y. Glioma induced alterations in fecal short-chain fatty acids and neurotransmitters. CNS Oncol 2020; 9:CNS57. [PMID: 32602743 PMCID: PMC7341178 DOI: 10.2217/cns-2020-0007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: To explore fecal short-chain fatty acids and neurotransmitter alterations in a mouse-glioma model and glioma patients. Methods: Liquid chromatography-mass spectrometry and 16S rRNA-sequencing from fecal samples were performed to measure metabolite levels and taxa abundance in mice/humans. Mice underwent GL261 implantation with/without temozolomide. Glioma patients were compared with healthy controls. Results: Glioma altered several short-chain fatty acids and neurotransmitter levels. Reduced 5-hydroxyindoleaceic acid and norepinephrine levels were seen in mice and humans. Interestingly, temozolomide treatment abrogates the effects of glioma on fecal metabolites. Conclusion: Our findings demonstrate the interplay between glioma and the gut-brain axis. Further work is required to identify pathways within the gut-brain axis by which glioma influences and promotes the modulation of fecal metabolites and microbiome.
Collapse
Affiliation(s)
- Antonio Dono
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Anthony Patrizz
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ryan M McCormack
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nagireddy Putluri
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TTX 77030, USA
| | - Bhanu P Ganesh
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TTX 77030, USA
| | - Balveen Kaur
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Louise D McCullough
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TTX 77030, USA
| | - Leomar Y Ballester
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Memorial Hermann Hospital-TMC, Houston, TX 77030, USA
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Memorial Hermann Hospital-TMC, Houston, TX 77030, USA.,Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TTX 77030, USA
| |
Collapse
|
43
|
UBE2T promotes glioblastoma invasion and migration via stabilizing GRP78 and regulating EMT. Aging (Albany NY) 2020; 12:10275-10289. [PMID: 32491994 PMCID: PMC7346020 DOI: 10.18632/aging.103239] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/31/2020] [Indexed: 01/10/2023]
Abstract
Glioblastoma (GBM) generally has a dismal prognosis, and it is associated with a poor quality of life as the disease progresses. However, the development of effective therapies for GBM has been deficient. Ubiquitin-conjugating enzyme E2T (UBE2T) is a member of the E2 family in the ubiquitin-proteasome pathway and a vital regulator of tumour progression, but its role in GBM is unclear. In this study, we aimed to clarify the role of UBE2T in GBM. Bioinformatics analysis identified UBE2T as an independent risk factor for gliomas. Immunohistochemistry was used to measure UBE2T expression in GBM and normal tissue samples obtained from patients with GBM. The effects of UBE2T on GBM cell invasion and migration were analysed using the Transwell assay. BALB/c nude mice were used for the in vivo assays. Immunoblotting and immunoprecipitation were performed to determine the molecular mechanisms. UBE2T was highly expressed in GBM tissues, and its expression was linked to a poor prognosis. In vitro, depletion of UBE2T significantly suppressed cell invasion and migration. Moreover, UBE2T depletion suppressed the growth of GBM subcutaneous tumours in vivo. Further experiments revealed that UBE2T suppressed invasion and migration by regulating epithelial- mesenchymal transition (EMT) via stabilising GRP78 in GBM cells. We uncovered a novel UBE2T/GRP78/EMT regulatory axis that modulates the malignant progression and recurrence of GBM, indicating that the axis might be a valuable therapeutic target.
Collapse
|
44
|
Thompson EG, Sontheimer H. Acetylcholine Receptor Activation as a Modulator of Glioblastoma Invasion. Cells 2019; 8:cells8101203. [PMID: 31590360 PMCID: PMC6829263 DOI: 10.3390/cells8101203] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
Grade IV astrocytomas, or glioblastomas (GBMs), are the most common malignant primary brain tumor in adults. The median GBM patient survival of 12–15 months has remained stagnant, in spite of treatment strategies, making GBMs a tremendous challenge clinically. This is at least in part due to the complex interaction of GBM cells with the brain microenvironment and their tendency to aggressively infiltrate normal brain tissue. GBMs frequently invade supratentorial brain regions that are richly innervated by neurotransmitter projections, most notably acetylcholine (ACh). Here, we asked whether ACh signaling influences the biology of GBMs. We examined the expression and function of known ACh receptors (AChRs) in large GBM datasets, as well as, human GBM cell lines and patient-derived xenograft lines. Using RNA-Seq data from the “The Cancer Genome Atlas” (TCGA), we confirmed the expression of AChRs and demonstrated the functionality of these receptors in GBM cells with time-lapse calcium imaging. AChR activation did not alter cell proliferation or migration, however, it significantly increased cell invasion through complex extracellular matrices. This was due to the enhanced activity of matrix metalloproteinase-9 (MMP-9) from GBM cells, which we found to be dependent on an intracellular calcium-dependent mechanism. Consistent with these findings, AChRs were significantly upregulated in regions of GBM infiltration in situ (Ivy Glioblastoma Atlas Project) and elevated expression of muscarinic AChR M3 correlated with reduced patient survival (TCGA). Data from the Repository for Molecular Brain Neoplasia Data (REMBRANDT) dataset also showed the co-expression of choline transporters, choline acetyltransferase, and vesicular acetylcholine transporters, suggesting that GBMs express all the proteins required for ACh synthesis and release. These findings identify ACh as a modulator of GBM behavior and posit that GBMs may utilize ACh as an autocrine signaling molecule.
Collapse
Affiliation(s)
- Emily G Thompson
- Glial Biology in Health, Disease and Cancer Center, Fralin Biomedical Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA.
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Harald Sontheimer
- Glial Biology in Health, Disease and Cancer Center, Fralin Biomedical Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA.
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA.
| |
Collapse
|
45
|
A Novel Multi-Target Small Molecule, LCC-09, Inhibits Stemness and Therapy-Resistant Phenotypes of Glioblastoma Cells by Increasing miR-34a and Deregulating the DRD4/Akt/mTOR Signaling Axis. Cancers (Basel) 2019; 11:cancers11101442. [PMID: 31561595 PMCID: PMC6826618 DOI: 10.3390/cancers11101442] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/17/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022] Open
Abstract
The management of glioblastomas (GBMs) is challenged by the development of therapeutic resistance and early disease recurrence, despite multi-modal therapy. This may be attributed to the presence of glioma stem cells (GSCs) which are known to survive radio- and chemotherapy, by circumventing death signals and inducing cell re-population. Recent findings suggest GSCs may be enriched by certain treatment modality. These necessitate the development of novel therapeutics capable of targeting GBM cell plasticity and therapy-resistant GSCs. Here, aided by computer-assisted structure characterization and target identification, we predicted that a novel 5-(2′,4′-difluorophenyl)-salicylanilide derivative, LCC-09, could target dopamine receptors and oncogenic markers implicated in GBMs. Bioinformatics data have indicated that dopamine receptor (DRD) 2, DRD4, CD133 and Nestin were elevated in GBM clinical samples and correlated to TMZ (Temozolomide) resistance and increased ALDH (Aldehyde dehydrogenase) activity (3.5–8.9%) as well as enhanced (2.1–2.4-fold) neurosphere formation efficiency in U87MG and D54MG GBM cell lines. In addition, TMZ-resistant GSC phenotype was associated with up-regulated DRD4, Akt, mTOR, β-catenin, CDK6, NF-κB and Erk1/2 expression. LCC-09 alone, or combined with TMZ, suppressed the tumorigenic and stemness traits of TMZ-resistant GBM cells while concomitantly down-regulating DRD4, Akt, mTOR, β-catenin, Erk1/2, NF-κB, and CDK6 expression. Notably, LCC-09-mediated anti-GBM/GSC activities were associated with the re-expression of tumor suppressor miR-34a and reversal of TMZ-resistance, in vitro and in vivo. Collectively, these data lay the foundation for further exploration of the clinical feasibility of administering LCC-09 as single-agent or combinatorial therapy for patients with TMZ-resistant GBMs.
Collapse
|
46
|
Role of monoamine-oxidase-A-gene variation in the development of glioblastoma in males: a case control study. J Neurooncol 2019; 145:287-294. [PMID: 31556016 PMCID: PMC6856259 DOI: 10.1007/s11060-019-03294-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/16/2019] [Indexed: 12/23/2022]
Abstract
Background The Mono-amine oxidase-A (MAO-A) enzyme is involved in the degradation and regulation of catecholamines such as serotonin, dopamine, epinephrine and nor-epinephrine. Preclinical studies suggest that this enzyme may contribute to an environment favorable for growth of malignant glioma. The MAO-A gene is located on the X-chromosome and has at least one functional genetic polymorphism. The aim of the present study was to explore possible effects of MAO-A genotype on development of glioblastoma in males. Methods Genotypes for 437 glioma cases and 876 population-based controls from the Swedish Glioma International Case–Control study (GICC) were compared. We analyzed the germline DNA using the Illumina Oncoarray. We selected seven single nucleotide polymorphisms (SNPs) located in the MAO-A gene, and imputed genotypes based on data from the 1000 genomes project. We used 1579 male glioblastoma cases and 1875 controls comprising the whole GICC cohort for subsequent validation of findings. Results The rs144551722 SNP was a significant predictor of development of glioblastoma in males (p-value = 0.0056) but not in females even after correction for multiple testing. We conducted haplotype analysis to confirm an association between MAO-A gene and risk of glioblastoma (p-value = 0.016). We found similar results in the validation sample. Conclusions These results suggest the possibility of a role for the MAO-A enzyme and the MAO-A gene in the development of glioblastoma in males.
Collapse
|
47
|
Li L, Liu X, Ma X, Deng X, Ji T, Hu P, Wan R, Qiu H, Cui D, Gao L. Identification of key candidate genes and pathways in glioblastoma by integrated bioinformatical analysis. Exp Ther Med 2019; 18:3439-3449. [PMID: 31602219 PMCID: PMC6777220 DOI: 10.3892/etm.2019.7975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM), characterized by high morbidity and mortality, is one of the most common lethal diseases worldwide. To identify the molecular mechanisms that contribute to the development of GBM, three cohort profile datasets (GSE50161, GSE90598 and GSE104291) were integrated and thoroughly analyzed; these datasets included 57 GBM cases and 22 cases of normal brain tissue. The current study identified differentially expressed genes (DEGs), and analyzed potential candidate genes and pathways. Additionally, a DEGs-associated protein-protein interaction (PPI) network was established for further investigation. Then, the hub genes associated with prognosis were identified using a Kaplan-Meier analysis based on The Cancer Genome Atlas database. Firstly, the current study identified 378 consistent DEGs (240 upregulated and 138 downregulated). Secondly, a cluster analysis of the DEGs was performed based on functions of the DEGs and signaling pathways were analyzed using the enrichment analysis tool on DAVID. Thirdly, 245 DEGs were identified using PPI network analysis. Among them, two co-expression modules comprising of 30 and 27 genes, respectively, and 35 hub genes were identified using Cytoscape MCODE. Finally, Kaplan-Meier analysis of the hub genes revealed that the increased expression of calcium-binding protein 1 (CABP1) was negatively associated with relapse-free survival. To summarize, all enriched Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways may participate in mechanisms underlying GBM occurrence and progression, however further studies are required. CABP1 may be a key gene associated with the biological process of GBM development and may be involved in a crucial mechanism of GBM progression.
Collapse
Affiliation(s)
- Lei Li
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Xiaohui Liu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Xiaoye Ma
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Xianyu Deng
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Tao Ji
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Pingping Hu
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Ronghao Wan
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Huijia Qiu
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Daming Cui
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China.,Department of Neurosurgery, Ninghai First Hospital, Ningbo, Zhejiang 315600, P.R. China
| | - Liang Gao
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China.,Department of Neurosurgery, Ninghai First Hospital, Ningbo, Zhejiang 315600, P.R. China
| |
Collapse
|
48
|
Pediatric and adult H3 K27M-mutant diffuse midline glioma treated with the selective DRD2 antagonist ONC201. J Neurooncol 2019; 145:97-105. [PMID: 31456142 DOI: 10.1007/s11060-019-03271-3] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND H3 K27M-mutant diffuse midline glioma is a fatal malignancy with no proven medical therapies. The entity predominantly occurs in children and young adults. ONC201 is a small molecule selective antagonist of dopamine receptor D2/3 (DRD2/3) with an exceptional safety profile. Following up on a durable response in the first H3 K27M-mutant diffuse midline glioma patient who received ONC201 (NCT02525692), an expanded access program was initiated. METHODS Patients with H3 K27M-mutant gliomas who received at least prior radiation were eligible. Patients with leptomeningeal spread were excluded. All patients received open-label ONC201 orally once every week. Safety, radiographic assessments, and overall survival were regularly assessed at least every 8 weeks by investigators. As of August 2018, a total of 18 patients with H3 K27M-mutant diffuse midline glioma or DIPG were enrolled to single patient expanded access ONC201 protocols. Among the 18 patients: seven adult (> 20 years old) and seven pediatric (< 20 years old) patients initiated ONC201 with recurrent disease and four pediatric patients initiated ONC201 following radiation, but prior to disease recurrence. FINDINGS Among the 14 patients with recurrent disease prior to initiation of ONC201, median progression-free survival is 14 weeks and median overall survival is 17 weeks. Three adults among the 14 recurrent patients remain on treatment progression-free with a median follow up of 49.6 (range 41-76.1) weeks. Among the 4 pediatric patients who initiated adjuvant ONC201 following radiation, two DIPG patients remain progression-free for at least 53 and 81 weeks. Radiographic regressions, including a complete response, were reported by investigators in a subset of patients with thalamic and pontine gliomas, along with improvements in disease-associated neurological symptoms. INTERPRETATION The clinical outcomes and radiographic responses in these patients provide the preliminary, and initial clinical proof-of-concept for targeting H3 K27M-mutant diffuse midline glioma with ONC201, regardless of age or location, providing rationale for robust clinical testing of the agent.
Collapse
|
49
|
Stein MN, Malhotra J, Tarapore RS, Malhotra U, Silk AW, Chan N, Rodriguez L, Aisner J, Aiken RD, Mayer T, Haffty BG, Newman JH, Aspromonte SM, Bommareddy PK, Estupinian R, Chesson CB, Sadimin ET, Li S, Medina DJ, Saunders T, Frankel M, Kareddula A, Damare S, Wesolowsky E, Gabel C, El-Deiry WS, Prabhu VV, Allen JE, Stogniew M, Oster W, Bertino JR, Libutti SK, Mehnert JM, Zloza A. Safety and enhanced immunostimulatory activity of the DRD2 antagonist ONC201 in advanced solid tumor patients with weekly oral administration. J Immunother Cancer 2019; 7:136. [PMID: 31118108 PMCID: PMC6532211 DOI: 10.1186/s40425-019-0599-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/23/2019] [Indexed: 11/10/2022] Open
Abstract
Background ONC201 is a small molecule antagonist of DRD2, a G protein-coupled receptor overexpressed in several malignancies, that has prolonged antitumor efficacy and immunomodulatory properties in preclinical models. The first-in-human trial of ONC201 previously established a recommended phase II dose (RP2D) of 625 mg once every three weeks. Here, we report the results of a phase I study that evaluated the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of weekly ONC201. Methods Patients ≥ 18 years old with an advanced solid tumor refractory to standard treatment were enrolled. Dose escalation proceeded with a 3 + 3 design from 375 mg to 625 mg of ONC201. One cycle, also the dose-limiting toxicity (DLT) window, was 21 days. The primary endpoint was to determine the RP2D of weekly ONC201, which was confirmed in an 11-patient dose expansion cohort. Results Twenty patients were enrolled: three at 375 mg and 17 at 625 mg of ONC201. The RP2D was defined as 625 mg with no DLT, treatment discontinuation, or dose modifications due to drug-related toxicity. PK profiles were consistent with every-three-week dosing and similar between the first and fourth dose. Serum prolactin and caspase-cleaved cytokeratin-18 induction were detected, along with intratumoral integrated stress response activation and infiltration of granzyme B+ Natural Killer cells. Induction of immune cytokines and effectors was higher in patients who received ONC201 once weekly versus once every three weeks. Stable disease of > 6 months was observed in several prostate and endometrial cancer patients. Conclusions Weekly, oral ONC201 is well-tolerated and results in enhanced immunostimulatory activity that warrants further investigation. Trial registration NCT02250781 (Oral ONC201 in Treating Patients With Advanced Solid Tumors), NCT02324621 (Continuation of Oral ONC201 in Treating Patients With Advanced Solid Tumors). Electronic supplementary material The online version of this article (10.1186/s40425-019-0599-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mark N Stein
- Division of Hematology/Oncology, Columbia University Medical Center, New York, NY, USA.,Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Jyoti Malhotra
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | | | - Usha Malhotra
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Ann W Silk
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Dermatology and Department of Medicine, Division of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nancy Chan
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Lorna Rodriguez
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Joseph Aisner
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Robert D Aiken
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Tina Mayer
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Bruce G Haffty
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Jenna H Newman
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Salvatore M Aspromonte
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Praveen K Bommareddy
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Ricardo Estupinian
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Charles B Chesson
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Evita T Sadimin
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Shengguo Li
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Daniel J Medina
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Tracie Saunders
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Melissa Frankel
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Aparna Kareddula
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Sherrie Damare
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Elayne Wesolowsky
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Christian Gabel
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Wafik S El-Deiry
- Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | | | | | | | | | - Joseph R Bertino
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Steven K Libutti
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Janice M Mehnert
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| | - Andrew Zloza
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA. .,Division of Hematology, Oncology, and Cell Therapy, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
50
|
Activation of Dopamine Receptor 2 Prompts Transcriptomic and Metabolic Plasticity in Glioblastoma. J Neurosci 2019; 39:1982-1993. [PMID: 30651332 DOI: 10.1523/jneurosci.1589-18.2018] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 12/17/2018] [Accepted: 12/28/2018] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive and lethal tumor types. Evidence continues to accrue indicating that the complex relationship between GBM and the brain microenvironment contributes to this malignant phenotype. However, the interaction between GBM and neurotransmitters, signaling molecules involved in neuronal communication, remains incompletely understood. Here we examined, using human patient-derived xenograft lines, how the monoamine dopamine influences GBM cells. We demonstrate that GBM cells express dopamine receptor 2 (DRD2), with elevated expression in the glioma-initiating cell (GIC) population. Stimulation of DRD2 caused a neuron-like hyperpolarization exclusively in GICs. In addition, long-term activation of DRD2 heightened the sphere-forming capacity of GBM cells, as well as tumor engraftment efficiency in both male and female mice. Mechanistic investigation revealed that DRD2 signaling activates the hypoxia response and functionally alters metabolism. Finally, we found that GBM cells synthesize and secrete dopamine themselves, suggesting a potential autocrine mechanism. These results identify dopamine signaling as a potential therapeutic target in GBM and further highlight neurotransmitters as a key feature of the pro-tumor microenvironment.SIGNIFICANCE STATEMENT This work offers critical insight into the role of the neurotransmitter dopamine in the progression of GBM. We show that dopamine induces specific changes in the state of tumor cells, augmenting their growth and shifting them to a more stem-cell like state. Further, our data illustrate that dopamine can alter the metabolic behavior of GBM cells, increasing glycolysis. Finally, this work demonstrates that GBM cells, including tumor samples from patients, can synthesize and secrete dopamine, suggesting an autocrine signaling process underlying these results. These results describe a novel connection between neurotransmitters and brain cancer, further highlighting the critical influence of the brain milieu on GBM.
Collapse
|