1
|
Freire NH, Herlinger AL, Vanini J, Dalmolin M, Fernandes MAC, Nör C, Ramaswamy V, de Farias CB, Brunetto AT, Brunetto AL, Gregianin LJ, Jaeger MDC, Taylor MD, Roesler R. Modulation of Stemness and Differentiation Regulators by Valproic Acid in Medulloblastoma Neurospheres. Cells 2025; 14:72. [PMID: 39851500 PMCID: PMC11763699 DOI: 10.3390/cells14020072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
Changes in epigenetic processes such as histone acetylation are proposed as key events influencing cancer cell function and the initiation and progression of pediatric brain tumors. Valproic acid (VPA) is an antiepileptic drug that acts partially by inhibiting histone deacetylases (HDACs) and could be repurposed as an epigenetic anticancer therapy. Here, we show that VPA reduced medulloblastoma (MB) cell viability and led to cell cycle arrest. These effects were accompanied by enhanced H3K9 histone acetylation (H3K9ac) and decreased expression of the MYC oncogene. VPA impaired the expansion of MB neurospheres enriched in stemness markers and reduced MYC while increasing TP53 expression in these neurospheres. In addition, VPA induced morphological changes consistent with neuronal differentiation and the increased expression of differentiation marker genes TUBB3 and ENO2. The expression of stemness genes SOX2, NES, and PRTG was differentially affected by VPA in MB cells with different TP53 status. VPA increased H3K9 occupancy of the promoter region of TP53. Among the genes regulated by VPA, the stemness regulators MYC and NES showed an association with patient survival in specific MB subgroups. Our results indicate that VPA may exert antitumor effects in MB by influencing histone acetylation, which may result in the modulation of stemness, neuronal differentiation, and the expression of genes associated with patient prognosis in specific molecular subgroups. Importantly, the actions of VPA in MB cells and neurospheres include a reduction in the expression of MYC and an increase in TP53.
Collapse
Affiliation(s)
- Natália Hogetop Freire
- Children’s Cancer Institute (ICI), Porto Alegre 90620-110, RS, Brazil
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
| | - Alice Laschuk Herlinger
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
| | - Julia Vanini
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
| | - Matheus Dalmolin
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
- InovAI Lab, nPITI/IMD, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
- Bioinformatics Multidisciplinary Environment (BioME), Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| | - Marcelo A. C. Fernandes
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
- InovAI Lab, nPITI/IMD, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
- Bioinformatics Multidisciplinary Environment (BioME), Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
- Department of Computer Engineering and Automation, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| | - Carolina Nör
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Vijay Ramaswamy
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Caroline Brunetto de Farias
- Children’s Cancer Institute (ICI), Porto Alegre 90620-110, RS, Brazil
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
| | - André Tesainer Brunetto
- Children’s Cancer Institute (ICI), Porto Alegre 90620-110, RS, Brazil
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
| | - Algemir Lunardi Brunetto
- Children’s Cancer Institute (ICI), Porto Alegre 90620-110, RS, Brazil
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
| | - Lauro José Gregianin
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
- Department of Pediatrics, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- Pediatric Oncology Service, Clinical Hospital, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
| | - Mariane da Cunha Jaeger
- Children’s Cancer Institute (ICI), Porto Alegre 90620-110, RS, Brazil
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
| | - Michael D. Taylor
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Texas Children’s Cancer and Hematology Center, Houston, TX 77030, USA
- Department of Pediatrics—Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neurosurgery, Texas Children’s Hospital, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Hematology-Oncology Section, Texas Children’s Cancer Center, Houston, TX 77030, USA
| | - Rafael Roesler
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology-INCT BioOncoPed, Porto Alegre 90035-003, RS, Brazil
- Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
| |
Collapse
|
2
|
Cristalli C, Scotlandi K. Targeting DNA Methylation Machinery in Pediatric Solid Tumors. Cells 2024; 13:1209. [PMID: 39056791 PMCID: PMC11275080 DOI: 10.3390/cells13141209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
DNA methylation is a key epigenetic regulatory mechanism that plays a critical role in a variety of cellular processes, including the regulation of cell fate during development, maintenance of cell identity, and genome stability. DNA methylation is tightly regulated by enzymatic reactions and its deregulation plays an important role in the development of cancer. Specific DNA methylation alterations have been found in pediatric solid tumors, providing new insights into the development of these tumors. In addition, DNA methylation profiles have greatly contributed to tune the diagnosis of pediatric solid tumors and to define subgroups of patients with different risks of progression, leading to the reduction in unwanted toxicity and the improvement of treatment efficacy. This review highlights the dysregulated DNA methylome in pediatric solid tumors and how this information provides promising targets for epigenetic therapies, particularly inhibitors of DNMT enzymes (DNMTis). Opportunities and limitations are considered, including the ability of DNMTis to induce viral mimicry and immune signaling by tumors. Besides intrinsic action against cancer cells, DNMTis have the potential to sensitize immune-cold tumors to immunotherapies and may represent a remarkable option to improve the treatment of challenging pediatric solid tumors.
Collapse
Affiliation(s)
- Camilla Cristalli
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy
| |
Collapse
|
3
|
Gorini F, Miceli M, de Antonellis P, Amente S, Zollo M, Ferrucci V. Epigenetics and immune cells in medulloblastoma. Front Genet 2023; 14:1135404. [PMID: 36968588 PMCID: PMC10036437 DOI: 10.3389/fgene.2023.1135404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Medulloblastoma (MB) is a highly malignant childhood tumor of the cerebellum. Transcriptional and epigenetic signatures have classified MB into four molecular subgroups, further stratified into biologically different subtypes with distinct somatic copy-number aberrations, driver genes, epigenetic alterations, activated pathways, and clinical outcomes. The brain tumor microenvironment (BTME) is of importance to regulate a complex network of cells, including immune cells, involved in cancer progression in brain malignancies. MB was considered with a “cold” immunophenotype due to the low influx of immune cells across the blood brain barrier (BBB). Recently, this assumption has been reconsidered because of the identification of infiltrating immune cells showing immunosuppressive phenotypes in the BTME of MB tumors. Here, we are providing a comprehensive overview of the current status of epigenetics alterations occurring during cancer progression with a description of the genomic landscape of MB by focusing on immune cells within the BTME. We further describe how new immunotherapeutic approaches could influence concurring epigenetic mechanisms of the immunosuppressive cells in BTME. In conclusion, the modulation of these molecular genetic complexes in BTME during cancer progression might enhance the therapeutic benefit, thus firing new weapons to fight MB.
Collapse
Affiliation(s)
- Francesca Gorini
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
| | - Marco Miceli
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| | - Pasqualino de Antonellis
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
| | - Stefano Amente
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- DAI Medicina di Laboratorio e Trasfusionale, ‘AOU Federico II Policlinico, Naples, Italy
| | - Veronica Ferrucci
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples, Naples, Italy
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- *Correspondence: Veronica Ferrucci,
| |
Collapse
|
4
|
Skitchenko R, Dinikina Y, Smirnov S, Krapivin M, Smirnova A, Morgacheva D, Artomov M. Case report: Somatic mutations in microtubule dynamics-associated genes in patients with WNT-medulloblastoma tumors. Front Oncol 2023; 12:1085947. [PMID: 36713498 PMCID: PMC9877404 DOI: 10.3389/fonc.2022.1085947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/07/2022] [Indexed: 01/14/2023] Open
Abstract
Medulloblastoma (MB) is the most common pediatric brain tumor which accounts for about 20% of all pediatric brain tumors and 63% of intracranial embryonal tumors. MB is considered to arise from precursor cell populations present during an early brain development. Most cases (~70%) of MB occur at the age of 1-4 and 5-9, but are also infrequently found in adults. Total annual frequency of pediatric tumors is about 5 cases per 1 million children. WNT-subtype of MB is characterized by a high probability of remission, with a long-term survival rate of about 90%. However, in some rare cases there may be increased metastatic activity, which dramatically reduces the likelihood of a favorable outcome. Here we report two cases of MB with a histological pattern consistent with desmoplastic/nodular (DP) and classic MB, and genetically classified as WNT-MB. Both cases showed putative causal somatic protein truncating mutations identified in microtubule-associated genes: ARID2, TUBB4A, and ANK3.
Collapse
Affiliation(s)
- Rostislav Skitchenko
- Almazov National Medical Research Centre, St. Petersburg, Russia,Computer Technologies Laboratory, ITMO University, St. Petersburg, Russia
| | - Yulia Dinikina
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Sergey Smirnov
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Mikhail Krapivin
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Anna Smirnova
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Daria Morgacheva
- Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Mykyta Artomov
- Almazov National Medical Research Centre, St. Petersburg, Russia,Computer Technologies Laboratory, ITMO University, St. Petersburg, Russia,The Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, United States,Department of Pediatrics, Ohio State University, Columbus, OH, United States,*Correspondence: Mykyta Artomov,
| |
Collapse
|
5
|
G9a/EHMT2 is a Potential Prognostic Biomarker and Molecular Target in SHH Medulloblastoma. Neuromolecular Med 2022; 24:392-398. [PMID: 35113321 DOI: 10.1007/s12017-022-08702-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/16/2022] [Indexed: 10/19/2022]
Abstract
Changes in epigenetic programming are associated with cancer development during childhood. Components of the epigenetic machinery involved in normal embryonic development and hijacked by pediatric cancers include enzymes mediating post-translational modifications of DNA and histones that regulate chromatin structure, such as histone methyltransferases (HMTs). Overexpression of the HMT G9a (euchromatic histone lysine methyltransferase 2, EHMT2) has been described in several cancer types. Medulloblastoma (MB), the main type of malignant brain tumor afflicting children, is currently classified into four molecular subgroups. Here, we show that expression level of the G9a/Ehmt2 gene is higher in MB tumors belonging to the SHH, Group 3, and Group 4 subgroups, compared to Wnt tumors. Remarkably, high G9a expression was significantly associated with shorter overall survival in MB patients. We also present evidence that G9a inhibition dose-dependently reduces MB cell viability. Our findings suggest that higher transcription of G9a may be a predictor of poor prognosis in patients with SHH MB, and that inhibiting G9a activity can display antitumor effects in MB.
Collapse
|
6
|
EHMT2/G9a as an Epigenetic Target in Pediatric and Adult Brain Tumors. Int J Mol Sci 2021; 22:ijms222011292. [PMID: 34681949 PMCID: PMC8539543 DOI: 10.3390/ijms222011292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/09/2021] [Indexed: 02/08/2023] Open
Abstract
Epigenetic mechanisms, including post-translational modifications of DNA and histones that influence chromatin structure, regulate gene expression during normal development and are also involved in carcinogenesis and cancer progression. The histone methyltransferase G9a (euchromatic histone lysine methyltransferase 2, EHMT2), which mostly mediates mono- and dimethylation by histone H3 lysine 9 (H3K9), influences gene expression involved in embryonic development and tissue differentiation. Overexpression of G9a has been observed in several cancer types, and different classes of G9a inhibitors have been developed as potential anticancer agents. Here, we review the emerging evidence suggesting the involvement of changes in G9a activity in brain tumors, namely glioblastoma (GBM), the main type of primary malignant brain cancer in adults, and medulloblastoma (MB), the most common type of malignant brain cancer in children. We also discuss the role of G9a in neuroblastoma (NB) and the drug development of G9a inhibitors.
Collapse
|
7
|
Epigenetic-Based Therapy-A Prospective Chance for Medulloblastoma Patients' Recovery. Int J Mol Sci 2021; 22:ijms22094925. [PMID: 34066495 PMCID: PMC8124462 DOI: 10.3390/ijms22094925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 12/25/2022] Open
Abstract
Medulloblastoma (MB) is one of the most frequent and malignant brain tumors in children. The prognosis depends on the advancement of the disease and the patient's age. Current therapies, which include surgery, chemotherapy, and irradiation, despite being quite effective, cause significant side effects that influence the central nervous system's function and cause neurocognitive deficits. Therefore, they substantially lower the quality of life, which is especially severe in a developing organism. Thus, there is a need for new therapies that are less toxic and even more effective. Recently, knowledge about the epigenetic mechanisms that are responsible for medulloblastoma development has increased. Epigenetics is a phenomenon that influences gene expression but can be easily modified by external factors. The best known epigenetic mechanisms are histone modifications, DNA methylation, or noncoding RNAs actions. Epigenetic mechanisms comprehensively explain the complex phenomena of carcinogenesis. At the same time, they seem to be a potential key to treating medulloblastoma with fewer complications than past therapies. This review presents the currently known epigenetic mechanisms that are involved in medulloblastoma pathogenesis and the potential therapies that use epigenetic traits to cure medulloblastoma while maintaining a good quality of life and ensuring a higher median overall survival rate.
Collapse
|
8
|
Amatruda JF. Modeling the developmental origins of pediatric cancer to improve patient outcomes. Dis Model Mech 2021; 14:14/2/dmm048930. [PMID: 33619212 PMCID: PMC7927656 DOI: 10.1242/dmm.048930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In the treatment of children and adolescents with cancer, multimodal approaches combining surgery, chemotherapy and radiation can cure most patients, but may cause lifelong health problems in survivors. Current therapies only modestly reflect increased knowledge about the molecular mechanisms of these cancers. Advances in next-generation sequencing have provided unprecedented cataloging of genetic aberrations in tumors, but understanding how these genetic changes drive cellular transformation, and how they can be effectively targeted, will require multidisciplinary collaboration and preclinical models that are truly representative of the in vivo environment. Here, I discuss some of the key challenges in pediatric cancer from my perspective as a physician-scientist, and touch on some promising new approaches that have the potential to transform our understanding of these diseases. Summary: This Perspective discusses the special features that make it challenging to develop new therapies for pediatric cancers, and the ways in which collaboration centered on improved models can meet these challenges.
Collapse
Affiliation(s)
- James F Amatruda
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| |
Collapse
|
9
|
Medulloblastoma drugs in development: Current leads, trials and drawbacks. Eur J Med Chem 2021; 215:113268. [PMID: 33636537 DOI: 10.1016/j.ejmech.2021.113268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 12/14/2022]
Abstract
Medulloblastoma (MB) is the most common malignant brain tumor in children. Current treatment for MB includes surgical resection, radiotherapy and chemotherapy. Despite significant progress in its management, a portion of children relapse and tumor recurrence carries a poor prognosis. Based on their molecular and clinical characteristics, MB patients are clinically classified into four groups: Wnt, Hh, Group 3, and Group 4. With our increased understanding of relevant molecular pathways disrupted in MB, the development of targeted therapies for MB has also increased. Targeted drugs have shown unique privileges over traditional cytotoxic therapies in balancing efficacy and toxicity, with many of them approved and widely used clinically. The aim of this review is to present the recent progress on targeted chemotherapies for the treatment of all classes of MB.
Collapse
|
10
|
Mueller T, Stucklin ASG, Postlmayr A, Metzger S, Gerber N, Kline C, Grotzer M, Nazarian J, Mueller S. Advances in Targeted Therapies for Pediatric Brain Tumors. Curr Treat Options Neurol 2020. [DOI: 10.1007/s11940-020-00651-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Abstract
Purpose of Review
Over the last years, our understanding of the molecular biology of pediatric brain tumors has vastly improved. This has led to more narrowly defined subgroups of these tumors and has created new potential targets for molecularly driven therapies. This review presents an overview of the latest advances and challenges of implementing targeted therapies into the clinical management of pediatric brain tumors, with a focus on gliomas, craniopharyngiomas, and medulloblastomas.
Recent Findings
Pediatric low-grade gliomas (pLGG) show generally a low mutational burden with the mitogen-activated protein kinase (MAPK) signaling presenting a key driver for these tumors. Direct inhibition of this pathway through BRAF and/or MEK inhibitors has proven to be a clinically relevant strategy. More recently, MEK and IL-6 receptor inhibitors have started to be evaluated in the treatment for craniopharyngiomas. Aside these low-grade tumors, pediatric high-grade gliomas (pHGG) and medulloblastomas exhibit substantially greater molecular heterogeneity with various and sometimes unknown tumor driver alterations. The clinical benefit of different targeted therapy approaches to interfere with altered signaling pathways and restore epigenetic dysregulation is undergoing active clinical testing. For these multiple pathway-driven tumors, combination strategies will most likely be required to achieve clinical benefit.
Summary
The field of pediatric neuro-oncology made tremendous progress with regard to improved diagnosis setting the stage for precision medicine approaches over the last decades. The potential of targeted therapies has been clearly demonstrated for a subset of pediatric brain tumors. However, despite clear response rates, questions of sufficient blood-brain barrier penetration, optimal dosing, treatment duration as well as mechanisms of resistance and how these can be overcome with potential combination strategies need to be addressed in future investigations. Along this line, it is critical for future trials to define appropriate endpoints to assess therapy responses as well as short and long-term toxicities in the growing and developing child.
Collapse
|
11
|
Alshawli AS, Wurdak H, Wood IC, Ladbury JE. Histone deacetylase inhibitors induce medulloblastoma cell death independent of HDACs recruited in REST repression complexes. Mol Genet Genomic Med 2020; 8:e1429. [PMID: 32720471 PMCID: PMC7549561 DOI: 10.1002/mgg3.1429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/05/2020] [Accepted: 07/02/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Repressor element 1-silencing transcription factor (REST) acts as a transcriptional repressor by recruiting several chromatin modifiers, including histone deacetylase (HDAC). Elevated REST expression in medulloblastoma has been associated with tumor progression nevertheless, the tumor shows high sensitivity to HDAC inhibitors (HDACi). However, the functional implications of REST and its requirement for HDACi-induced anti-cancer effects are not well understood. METHODS In this study, the expression of REST was evaluated across the medulloblastoma subgroups and subtypes using published gene expression data. Further, the expression of REST was modulated using the CRISPR/Cas9 knockout and shRNA knockdown in the Daoy medulloblastoma cell line. RESULTS The results of this study showed that the expression of REST is elevated in most medulloblastoma subgroups compared to the non-cancerous cerebellum. Blocking of REST expression resulted in increasing the expression of REST-regulated genes, a moderate decrease in the fraction of the cells in the S-phase, and reducing the cells' migration ability. However, REST deficiency did not lead to a marked decrease in the Daoy cell viability and sensitivity to HDACi. CONCLUSION The findings of this study indicate that REST is not essential for sustaining the proliferation/viability of the Daoy cells. It also revealed that the anti-proliferative effect of HDACi is independent of REST expression.
Collapse
Affiliation(s)
- Abdulelah S. Alshawli
- School of Biomedical SciencesFaculty of Biological SciencesUniversity of LeedsLeedsUK
| | - Heiko Wurdak
- Leeds Institute of Cancer and PathologyUniversity of LeedsSt James's University HospitalLeedsUK
| | - Ian C. Wood
- School of Biomedical SciencesFaculty of Biological SciencesUniversity of LeedsLeedsUK
| | - John E. Ladbury
- School of Molecular and Cellular BiologyFaculty of Biological SciencesUniversity of LeedsLeedsUK
| |
Collapse
|
12
|
Dual inhibitors of histone deacetylases and other cancer-related targets: A pharmacological perspective. Biochem Pharmacol 2020; 182:114224. [PMID: 32956642 DOI: 10.1016/j.bcp.2020.114224] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/01/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022]
Abstract
Epigenetic enzymes histone deacetylases (HDACs) are clinically validated anticancer drug targets which have been studied intensively in the past few decades. Although several drugs have been approved in this field, they are still limited to a subset of hematological malignancies (in particular T-cell lymphomas), with therapeutic potential not fully realized and the drug-resistance occurred after a certain period of use. To maximize the therapeutic potential of these classes of anticancer drugs, and to extend their application to solid tumors, numerous combination therapies containing an HDACi and an anticancer agent from other mechanisms are currently ongoing in clinical trials. Recently, dual targeting strategy comprising the HDACs component has emerged as an alternative approach for combination therapies. In this perspective, we intend to gather all HDACs-containing dual inhibitors related to cancer therapy published in literature since 2015, classify them into five categories based on targets' biological functions, and discuss the rationale why dual acting agents should work better than combinatorial therapies using two separate drugs. The article discusses the pharmacological aspects of these dual inhibitors, including in vitro biological activities, pharmacokinetic studies, in vivo efficacy studies, as well as available clinical trials. The review of the current status and advances should provide better analysis for future opportunities and challenges of this field.
Collapse
|
13
|
Mensà E, Guescini M, Giuliani A, Bacalini MG, Ramini D, Corleone G, Ferracin M, Fulgenzi G, Graciotti L, Prattichizzo F, Sorci L, Battistelli M, Monsurrò V, Bonfigli AR, Cardelli M, Recchioni R, Marcheselli F, Latini S, Maggio S, Fanelli M, Amatori S, Storci G, Ceriello A, Stocchi V, De Luca M, Magnani L, Rippo MR, Procopio AD, Sala C, Budimir I, Bassi C, Negrini M, Garagnani P, Franceschi C, Sabbatinelli J, Bonafè M, Olivieri F. Small extracellular vesicles deliver miR-21 and miR-217 as pro-senescence effectors to endothelial cells. J Extracell Vesicles 2020; 9:1725285. [PMID: 32158519 PMCID: PMC7048230 DOI: 10.1080/20013078.2020.1725285] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 01/26/2020] [Accepted: 01/29/2020] [Indexed: 12/17/2022] Open
Abstract
The role of epigenetics in endothelial cell senescence is a cutting-edge topic in ageing research. However, little is known of the relative contribution to pro-senescence signal propagation provided by microRNAs shuttled by extracellular vesicles (EVs) released from senescent cells. Analysis of microRNA and DNA methylation profiles in non-senescent (control) and senescent (SEN) human umbilical vein endothelial cells (HUVECs), and microRNA profiling of their cognate small EVs (sEVs) and large EVs demonstrated that SEN cells released a significantly greater sEV number than control cells. sEVs were enriched in miR-21-5p and miR-217, which target DNMT1 and SIRT1. Treatment of control cells with SEN sEVs induced a miR-21/miR-217-related impairment of DNMT1-SIRT1 expression, the reduction of proliferation markers, the acquisition of a senescent phenotype and a partial demethylation of the locus encoding for miR-21. MicroRNA profiling of sEVs from plasma of healthy subjects aged 40-100 years showed an inverse U-shaped age-related trend for miR-21-5p, consistent with senescence-associated biomarker profiles. Our findings suggest that miR-21-5p/miR-217 carried by SEN sEVs spread pro-senescence signals, affecting DNA methylation and cell replication.
Collapse
Affiliation(s)
- Emanuela Mensà
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Michele Guescini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | | | - Deborah Ramini
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Giacomo Corleone
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Manuela Ferracin
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Gianluca Fulgenzi
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Laura Graciotti
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | | | - Leonardo Sorci
- Department of Materials, Environmental Sciences and Urban Planning, Università Politecnica delle Marche, Ancona, Italy
| | - Michela Battistelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | | | | | - Maurizio Cardelli
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Ancona, Italy
| | - Rina Recchioni
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | | | - Silvia Latini
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Serena Maggio
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mirco Fanelli
- Department of Biomolecular Sciences, Molecular Pathology Laboratory “Paola”, University of Urbino Carlo Bo, Fano, Italy
| | - Stefano Amatori
- Department of Biomolecular Sciences, Molecular Pathology Laboratory “Paola”, University of Urbino Carlo Bo, Fano, Italy
| | - Gianluca Storci
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | | | - Vilberto Stocchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Maria De Luca
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, USA
| | - Luca Magnani
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Claudia Sala
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Iva Budimir
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Cristian Bassi
- Department of Morphology, Surgery & Experimental Medicine, and Laboratory for the Technologies of Advanced Therapies, Tecnopolo, University of Ferrara, Ferrara, Italy
| | - Massimo Negrini
- Department of Morphology, Surgery & Experimental Medicine, and Laboratory for the Technologies of Advanced Therapies, Tecnopolo, University of Ferrara, Ferrara, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden
- Personal Genomics S.r.l., Verona, Italy
| | - Claudio Franceschi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| |
Collapse
|