Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cancer Discov ; 2024 May 20.
Article in English | MEDLINE | ID: mdl-38767413

ABSTRACT

High-grade gliomas (HGG) are deadly diseases for both adult and pediatric patients. Recently, it has been shown that neuronal activity promotes progression of multiple subgroups of HGG. However, epigenetic mechanisms that govern this process remain elusive. Here we report that the chromatin remodeler CHD2 regulates neuron-glioma interactions in diffuse midline glioma (DMG) characterized by onco-histone H3.1K27M. Depletion of CHD2 in H3.1K27M DMG cells compromises cell viability and neuron-to-glioma synaptic connections in vitro, neuron-induced proliferation of H3.1K27M DMG cells in vitro and in vivo, activity-dependent calcium transients in vivo, and extends the survival of H3.1K27M DMG-bearing mice. Mechanistically, CHD2 coordinates with the transcription factor FOSL1 to control the expression of axon-guidance and synaptic genes in H3.1K27M DMG cells. Together, our study reveals a mechanism whereby CHD2 controls the intrinsic gene program of the H3.1K27M DMG subtype, which in turn regulates the tumor growth-promoting interactions of glioma cells with neurons.

2.
Nature ; 623(7986): 366-374, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37914930

ABSTRACT

The role of the nervous system in the regulation of cancer is increasingly appreciated. In gliomas, neuronal activity drives tumour progression through paracrine signalling factors such as neuroligin-3 and brain-derived neurotrophic factor1-3 (BDNF), and also through electrophysiologically functional neuron-to-glioma synapses mediated by AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) receptors4,5. The consequent glioma cell membrane depolarization drives tumour proliferation4,6. In the healthy brain, activity-regulated secretion of BDNF promotes adaptive plasticity of synaptic connectivity7,8 and strength9-15. Here we show that malignant synapses exhibit similar plasticity regulated by BDNF. Signalling through the receptor tropomyosin-related kinase B16 (TrkB) to CAMKII, BDNF promotes AMPA receptor trafficking to the glioma cell membrane, resulting in increased amplitude of glutamate-evoked currents in the malignant cells. Linking plasticity of glioma synaptic strength to tumour growth, graded optogenetic control of glioma membrane potential demonstrates that greater depolarizing current amplitude promotes increased glioma proliferation. This potentiation of malignant synaptic strength shares mechanistic features with synaptic plasticity17-22 that contributes to memory and learning in the healthy brain23-26. BDNF-TrkB signalling also regulates the number of neuron-to-glioma synapses. Abrogation of activity-regulated BDNF secretion from the brain microenvironment or loss of glioma TrkB expression robustly inhibits tumour progression. Blocking TrkB genetically or pharmacologically abrogates these effects of BDNF on glioma synapses and substantially prolongs survival in xenograft models of paediatric glioblastoma and diffuse intrinsic pontine glioma. Together, these findings indicate that BDNF-TrkB signalling promotes malignant synaptic plasticity and augments tumour progression.


Subject(s)
Adaptation, Physiological , Glioma , Neuronal Plasticity , Synapses , Animals , Child , Humans , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Proliferation , Disease Progression , Glioma/metabolism , Glioma/pathology , Glutamic Acid/metabolism , Neurons/cytology , Neurons/metabolism , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptors, AMPA/metabolism , Signal Transduction , Synapses/metabolism , Tumor Microenvironment , Optogenetics
3.
Nat Rev Neurosci ; 24(12): 733-746, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37857838

ABSTRACT

Experience sculpts brain structure and function. Activity-dependent modulation of the myelinated infrastructure of the nervous system has emerged as a dimension of adaptive change during childhood development and in adulthood. Myelination is a richly dynamic process, with neuronal activity regulating oligodendrocyte precursor cell proliferation, oligodendrogenesis and myelin structural changes in some axonal subtypes and in some regions of the nervous system. This myelin plasticity and consequent changes to conduction velocity and circuit dynamics can powerfully influence neurological functions, including learning and memory. Conversely, disruption of the mechanisms mediating adaptive myelination can contribute to cognitive impairment. The robust effects of neuronal activity on normal oligodendroglial precursor cells, a putative cellular origin for many forms of glioma, indicates that dysregulated or 'hijacked' mechanisms of myelin plasticity could similarly promote growth in this devastating group of brain cancers. Indeed, neuronal activity promotes the pathogenesis of many forms of glioma in preclinical models through activity-regulated paracrine factors and direct neuron-to-glioma synapses. This synaptic integration of glioma into neural circuits is central to tumour growth and invasion. Thus, not only do neuron-oligodendroglial interactions modulate neural circuit structure and function in the healthy brain, but neuron-glioma interactions also have important roles in the pathogenesis of glial malignancies.


Subject(s)
Glioma , Neurons , Humans , Neurons/physiology , Oligodendroglia/physiology , Myelin Sheath/physiology , Neuroglia/physiology
4.
Nature ; 619(7971): 844-850, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380778

ABSTRACT

The tumour microenvironment plays an essential role in malignancy, and neurons have emerged as a key component of the tumour microenvironment that promotes tumourigenesis across a host of cancers1,2. Recent studies on glioblastoma (GBM) highlight bidirectional signalling between tumours and neurons that propagates a vicious cycle of proliferation, synaptic integration and brain hyperactivity3-8; however, the identity of neuronal subtypes and tumour subpopulations driving this phenomenon is incompletely understood. Here we show that callosal projection neurons located in the hemisphere contralateral to primary GBM tumours promote progression and widespread infiltration. Using this platform to examine GBM infiltration, we identified an activity-dependent infiltrating population present at the leading edge of mouse and human tumours that is enriched for axon guidance genes. High-throughput, in vivo screening of these genes identified SEMA4F as a key regulator of tumourigenesis and activity-dependent progression. Furthermore, SEMA4F promotes the activity-dependent infiltrating population and propagates bidirectional signalling with neurons by remodelling tumour-adjacent synapses towards brain network hyperactivity. Collectively our studies demonstrate that subsets of neurons in locations remote to primary GBM promote malignant progression, and also show new mechanisms of glioma progression that are regulated by neuronal activity.


Subject(s)
Brain Neoplasms , Carcinogenesis , Glioma , Neurons , Tumor Microenvironment , Humans , Brain/pathology , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Carcinogenesis/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Glioblastoma/pathology , Glioblastoma/physiopathology , Glioma/pathology , Glioma/physiopathology , Neurons/pathology , Cell Proliferation , Synapses , Disease Progression , Animals , Mice , Axons , Corpus Callosum/pathology , Neural Pathways
5.
bioRxiv ; 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36993539

ABSTRACT

The tumor microenvironment (TME) plays an essential role in malignancy and neurons have emerged as a key component of the TME that promotes tumorigenesis across a host of cancers. Recent studies on glioblastoma (GBM) highlight bi-directional signaling between tumors and neurons that propagates a vicious cycle of proliferation, synaptic integration, and brain hyperactivity; however, the identity of neuronal subtypes and tumor subpopulations driving this phenomenon are incompletely understood. Here we show that callosal projection neurons located in the hemisphere contralateral to primary GBM tumors promote progression and widespread infiltration. Using this platform to examine GBM infiltration, we identified an activity dependent infiltrating population present at the leading edge of mouse and human tumors that is enriched for axon guidance genes. High-throughput, in vivo screening of these genes identified Sema4F as a key regulator of tumorigenesis and activity-dependent infiltration. Furthermore, Sema4F promotes the activity-dependent infiltrating population and propagates bi-directional signaling with neurons by remodeling tumor adjacent synapses towards brain network hyperactivity. Collectively, our studies demonstrate that subsets of neurons in locations remote to primary GBM promote malignant progression, while revealing new mechanisms of tumor infiltration that are regulated by neuronal activity.

6.
Cell ; 185(16): 2846-2848, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35931016

ABSTRACT

Glioblastoma is a lethal, diffusely invasive brain cancer that is robustly regulated by the activity of the brain itself, in part through neuron-to-glioma synaptic communication. Venkataramani et al. have conceptually advanced understanding of glioblastoma interactions with neural circuits, demonstrating that conduction of electrochemical signals via neuron-to-glioma synapses drives glioma invasion.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/pathology , Glioma/pathology , Humans , Neoplasm Invasiveness/pathology , Neurons/pathology
7.
Cell ; 185(14): 2452-2468.e16, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35768006

ABSTRACT

COVID survivors frequently experience lingering neurological symptoms that resemble cancer-therapy-related cognitive impairment, a syndrome for which white matter microglial reactivity and consequent neural dysregulation is central. Here, we explored the neurobiological effects of respiratory SARS-CoV-2 infection and found white-matter-selective microglial reactivity in mice and humans. Following mild respiratory COVID in mice, persistently impaired hippocampal neurogenesis, decreased oligodendrocytes, and myelin loss were evident together with elevated CSF cytokines/chemokines including CCL11. Systemic CCL11 administration specifically caused hippocampal microglial reactivity and impaired neurogenesis. Concordantly, humans with lasting cognitive symptoms post-COVID exhibit elevated CCL11 levels. Compared with SARS-CoV-2, mild respiratory influenza in mice caused similar patterns of white-matter-selective microglial reactivity, oligodendrocyte loss, impaired neurogenesis, and elevated CCL11 at early time points, but after influenza, only elevated CCL11 and hippocampal pathology persisted. These findings illustrate similar neuropathophysiology after cancer therapy and respiratory SARS-CoV-2 infection which may contribute to cognitive impairment following even mild COVID.


Subject(s)
COVID-19 , Influenza, Human , Neoplasms , Animals , Humans , Influenza, Human/pathology , Mice , Microglia/pathology , Myelin Sheath , Neoplasms/pathology , SARS-CoV-2
8.
bioRxiv ; 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35043113

ABSTRACT

Survivors of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection frequently experience lingering neurological symptoms, including impairment in attention, concentration, speed of information processing and memory. This long-COVID cognitive syndrome shares many features with the syndrome of cancer therapy-related cognitive impairment (CRCI). Neuroinflammation, particularly microglial reactivity and consequent dysregulation of hippocampal neurogenesis and oligodendrocyte lineage cells, is central to CRCI. We hypothesized that similar cellular mechanisms may contribute to the persistent neurological symptoms associated with even mild SARS-CoV-2 respiratory infection. Here, we explored neuroinflammation caused by mild respiratory SARS-CoV-2 infection - without neuroinvasion - and effects on hippocampal neurogenesis and the oligodendroglial lineage. Using a mouse model of mild respiratory SARS-CoV-2 infection induced by intranasal SARS-CoV-2 delivery, we found white matter-selective microglial reactivity, a pattern observed in CRCI. Human brain tissue from 9 individuals with COVID-19 or SARS-CoV-2 infection exhibits the same pattern of prominent white matter-selective microglial reactivity. In mice, pro-inflammatory CSF cytokines/chemokines were elevated for at least 7-weeks post-infection; among the chemokines demonstrating persistent elevation is CCL11, which is associated with impairments in neurogenesis and cognitive function. Humans experiencing long-COVID with cognitive symptoms (48 subjects) similarly demonstrate elevated CCL11 levels compared to those with long-COVID who lack cognitive symptoms (15 subjects). Impaired hippocampal neurogenesis, decreased oligodendrocytes and myelin loss in subcortical white matter were evident at 1 week, and persisted until at least 7 weeks, following mild respiratory SARS-CoV-2 infection in mice. Taken together, the findings presented here illustrate striking similarities between neuropathophysiology after cancer therapy and after SARS-CoV-2 infection, and elucidate cellular deficits that may contribute to lasting neurological symptoms following even mild SARS-CoV-2 infection.

9.
Cell ; 181(7): 1445-1449, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32533917

ABSTRACT

The COVID19 crisis has magnified the issues plaguing academic science, but it has also provided the scientific establishment with an unprecedented opportunity to reset. Shoring up the foundation of academic science will require a concerted effort between funding agencies, universities, and the public to rethink how we support scientists, with a special emphasis on early career researchers.


Subject(s)
Career Mobility , Research Personnel/trends , Research/trends , Achievement , Biomedical Research , Humans , Research Personnel/education , Science/education , Science/trends , Universities
10.
Nature ; 573(7775): 539-545, 2019 09.
Article in English | MEDLINE | ID: mdl-31534222

ABSTRACT

High-grade gliomas are lethal brain cancers whose progression is robustly regulated by neuronal activity. Activity-regulated release of growth factors promotes glioma growth, but this alone is insufficient to explain the effect that neuronal activity exerts on glioma progression. Here we show that neuron and glioma interactions include electrochemical communication through bona fide AMPA receptor-dependent neuron-glioma synapses. Neuronal activity also evokes non-synaptic activity-dependent potassium currents that are amplified by gap junction-mediated tumour interconnections, forming an electrically coupled network. Depolarization of glioma membranes assessed by in vivo optogenetics promotes proliferation, whereas pharmacologically or genetically blocking electrochemical signalling inhibits the growth of glioma xenografts and extends mouse survival. Emphasizing the positive feedback mechanisms by which gliomas increase neuronal excitability and thus activity-regulated glioma growth, human intraoperative electrocorticography demonstrates increased cortical excitability in the glioma-infiltrated brain. Together, these findings indicate that synaptic and electrical integration into neural circuits promotes glioma progression.


Subject(s)
Brain/physiopathology , Electrical Synapses/pathology , Electrophysiological Phenomena , Glioma/physiopathology , Animals , Brain/cytology , Cell Membrane/pathology , Cell Proliferation , Gap Junctions/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Mice , Mice, Inbred NOD , Neurons/pathology , Optogenetics , Potassium/metabolism , Synaptic Transmission , Tumor Cells, Cultured
11.
Commun Biol ; 2: 156, 2019.
Article in English | MEDLINE | ID: mdl-31098401

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a lethal childhood brainstem tumour, with a quarter of patients harbouring somatic mutations in ACVR1, encoding the serine/threonine kinase ALK2. Despite being an amenable drug target, little has been done to-date to systematically evaluate the role of ACVR1 in DIPG, nor to screen currently available inhibitors in patient-derived tumour models. Here we show the dependence of DIPG cells on the mutant receptor, and the preclinical efficacy of two distinct chemotypes of ALK2 inhibitor in vitro and in vivo. We demonstrate the pyrazolo[1,5-a]pyrimidine LDN-193189 and the pyridine LDN-214117 to be orally bioavailable and well-tolerated, with good brain penetration. Treatment of immunodeprived mice bearing orthotopic xenografts of H3.3K27M, ACVR1R206H mutant HSJD-DIPG-007 cells with 25 mg/kg LDN-193189 or LDN-214117 for 28 days extended survival compared with vehicle controls. Development of ALK2 inhibitors with improved potency, selectivity and advantageous pharmacokinetic properties may play an important role in therapy for DIPG patients.


Subject(s)
Activin Receptors, Type I/genetics , Antineoplastic Agents/pharmacology , Brain Stem Neoplasms/drug therapy , Diffuse Intrinsic Pontine Glioma/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/metabolism , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Apoptosis/genetics , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/mortality , Brain Stem Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Child , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/mortality , Diffuse Intrinsic Pontine Glioma/pathology , Drug Administration Schedule , Drug Evaluation, Preclinical , Female , Gene Expression , High-Throughput Screening Assays , Humans , Mice , Mice, SCID , Mutation , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyridines/pharmacokinetics , Pyrimidines/pharmacokinetics , Survival Analysis , Xenograft Model Antitumor Assays
12.
Nat Med ; 24(8): 1204-1215, 2018 08.
Article in English | MEDLINE | ID: mdl-29967352

ABSTRACT

The failure to develop effective therapies for pediatric glioblastoma (pGBM) and diffuse intrinsic pontine glioma (DIPG) is in part due to their intrinsic heterogeneity. We aimed to quantitatively assess the extent to which this was present in these tumors through subclonal genomic analyses and to determine whether distinct tumor subpopulations may interact to promote tumorigenesis by generating subclonal patient-derived models in vitro and in vivo. Analysis of 142 sequenced tumors revealed multiple tumor subclones, spatially and temporally coexisting in a stable manner as observed by multiple sampling strategies. We isolated genotypically and phenotypically distinct subpopulations that we propose cooperate to enhance tumorigenicity and resistance to therapy. Inactivating mutations in the H4K20 histone methyltransferase KMT5B (SUV420H1), present in <1% of cells, abrogate DNA repair and confer increased invasion and migration on neighboring cells, in vitro and in vivo, through chemokine signaling and modulation of integrins. These data indicate that even rare tumor subpopulations may exert profound effects on tumorigenesis as a whole and may represent a new avenue for therapeutic development. Unraveling the mechanisms of subclonal diversity and communication in pGBM and DIPG will be an important step toward overcoming barriers to effective treatments.


Subject(s)
Brain Stem Neoplasms/pathology , Glioblastoma/pathology , Animals , Brain Stem Neoplasms/genetics , Carcinogenesis/pathology , Cell Separation , Child , Clone Cells , Genotype , Glioblastoma/genetics , Humans , Mice, Nude , Phenotype , Tumor Cells, Cultured
13.
J Pathol ; 244(2): 242-253, 2018 02.
Article in English | MEDLINE | ID: mdl-29160922

ABSTRACT

Testicular germ cell tumours (TGCTs) are the most frequent malignancy and cause of death from solid tumours in the 20- to 40-year age group. Although most cases show sensitivity to cis-platinum-based chemotherapy, this is associated with long-term toxicities and chemo-resistance. Roles for receptor tyrosine kinases other than KIT are largely unknown in TGCT. We therefore conducted a phosphoproteomic screen and identified the insulin growth factor receptor-1 (IGF1R) as both highly expressed and activated in TGCT cell lines representing the nonseminomatous subtype. IGF1R was also frequently expressed in tumour samples from patients with nonseminomas. Functional analysis of cell line models showed that long-term shRNA-mediated IGF1R silencing leads to apoptosis and complete ablation of nonseminoma cells with active IGF1R signalling. Cell lines with high levels of IGF1R activity also showed reduced AKT signalling in response to decreased IGF1R expression as well as sensitivity to the small-molecule IGF1R inhibitor NVP-AEW541. These results were in contrast to those in the seminoma cell line TCAM2 that lacked IGF1R signalling via AKT and was one of the two cell lines least sensitive to the IGF1R inhibitor. The dependence on IGF1R activity in the majority of nonseminomas parallels the known role of IGF signalling in the proliferation, migration, and survival of primordial germ cells, the putative cell of origin for TGCT. Upregulation of IGF1R expression and signalling was also found to contribute to acquired cisplatin resistance in an in vitro nonseminoma model, providing a rationale for targeting IGF1R in cisplatin-resistant disease. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Neoplasms, Germ Cell and Embryonal/drug therapy , Receptors, Somatomedin/metabolism , Signal Transduction/drug effects , Testicular Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Male , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
14.
Cancer Cell ; 32(4): 520-537.e5, 2017 10 09.
Article in English | MEDLINE | ID: mdl-28966033

ABSTRACT

We collated data from 157 unpublished cases of pediatric high-grade glioma and diffuse intrinsic pontine glioma and 20 publicly available datasets in an integrated analysis of >1,000 cases. We identified co-segregating mutations in histone-mutant subgroups including loss of FBXW7 in H3.3G34R/V, TOP3A rearrangements in H3.3K27M, and BCOR mutations in H3.1K27M. Histone wild-type subgroups are refined by the presence of key oncogenic events or methylation profiles more closely resembling lower-grade tumors. Genomic aberrations increase with age, highlighting the infant population as biologically and clinically distinct. Uncommon pathway dysregulation is seen in small subsets of tumors, further defining the molecular diversity of the disease, opening up avenues for biological study and providing a basis for functionally defined future treatment stratification.


Subject(s)
Brain Stem Neoplasms/genetics , Glioma/genetics , Histones/genetics , Mutation , Adolescent , Brain Stem Neoplasms/pathology , Cell Cycle Proteins/genetics , Child , Child, Preschool , DNA Topoisomerases, Type I/genetics , Exome , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Female , Gene Dosage , Glioma/pathology , Humans , Infant , Infant, Newborn , Male , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Young Adult
15.
Cancer Cell ; 31(5): 635-652.e6, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28434841

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a fatal pediatric cancer with limited therapeutic options. The majority of cases of DIPG exhibit a mutation in histone-3 (H3K27M) that results in oncogenic transcriptional aberrancies. We show here that DIPG is vulnerable to transcriptional disruption using bromodomain inhibition or CDK7 blockade. Targeting oncogenic transcription through either of these methods synergizes with HDAC inhibition, and DIPG cells resistant to HDAC inhibitor therapy retain sensitivity to CDK7 blockade. Identification of super-enhancers in DIPG provides insights toward the cell of origin, highlighting oligodendroglial lineage genes, and reveals unexpected mechanisms mediating tumor viability and invasion, including potassium channel function and EPH receptor signaling. The findings presented demonstrate transcriptional vulnerabilities and elucidate previously unknown mechanisms of DIPG pathobiology.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azepines/pharmacology , Brain Stem Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Phenylenediamines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Transcription, Genetic/drug effects , Triazoles/pharmacology , Animals , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/metabolism , Brain Stem Neoplasms/pathology , Cell Cycle Proteins , Cell Proliferation/drug effects , Chromatin Assembly and Disassembly/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Synergism , Female , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Histones/genetics , Histones/metabolism , Humans , Male , Mice, Inbred NOD , Mice, SCID , Mutation , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Panobinostat , Primary Cell Culture , RNA Interference , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Signal Transduction/drug effects , Time Factors , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Cyclin-Dependent Kinase-Activating Kinase
16.
Acta Neuropathol ; 130(6): 815-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26399631

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is the most severe paediatric solid tumour, with no significant therapeutic progress made in the past 50 years. Recent studies suggest that diffuse midline glioma, H3-K27M mutant, may comprise more than one biological entity. The aim of the study was to determine the clinical and biological variables that most impact their prognosis. Ninety-one patients with classically defined DIPG underwent a systematic stereotactic biopsy and were included in this observational retrospective study. Histone H3 genes mutations were assessed by immunochemistry and direct sequencing, whilst global gene expression profiling and chromosomal imbalances were determined by microarrays. A full description of the MRI findings at diagnosis and at relapse was integrated with the molecular profiling data and clinical outcome. All DIPG but one were found to harbour either a somatic H3-K27M mutation and/or loss of H3K27 trimethylation. We also discovered a novel K27M mutation in HIST2H3C, and a lysine-to-isoleucine substitution (K27I) in H3F3A, also creating a loss of trimethylation. Patients with tumours harbouring a K27M mutation in H3.3 (H3F3A) did not respond clinically to radiotherapy as well, relapsed significantly earlier and exhibited more metastatic recurrences than those in H3.1 (HIST1H3B/C). H3.3-K27M-mutated DIPG have a proneural/oligodendroglial phenotype and a pro-metastatic gene expression signature with PDGFRA activation, while H3.1-K27M-mutated tumours exhibit a mesenchymal/astrocytic phenotype and a pro-angiogenic/hypoxic signature supported by expression profiling and radiological findings. H3K27 alterations appear as the founding event in DIPG and the mutations in the two main histone H3 variants drive two distinct oncogenic programmes with potential specific therapeutic targets.


Subject(s)
Brain Stem Neoplasms/genetics , Glioma/genetics , Histones/genetics , Mutation , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/radiation effects , Brain Stem Neoplasms/diagnosis , Brain Stem Neoplasms/pathology , Brain Stem Neoplasms/radiotherapy , Child , Child, Preschool , Cohort Studies , Female , Glioma/diagnosis , Glioma/pathology , Glioma/radiotherapy , HeLa Cells , Humans , Male , Neurons/metabolism , Neurons/pathology , Neurons/radiation effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligodendroglia/radiation effects , Phenotype , Pons/metabolism , Pons/pathology , Pons/radiation effects , Pons/surgery , Prognosis
17.
Cancer Res ; 74(17): 4565-70, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25136070

ABSTRACT

Whole-genome sequencing studies have recently identified a quarter of cases of the rare childhood brainstem tumor diffuse intrinsic pontine glioma to harbor somatic mutations in ACVR1. This gene encodes the type I bone morphogenic protein receptor ALK2, with the residues affected identical to those that, when mutated in the germline, give rise to the congenital malformation syndrome fibrodysplasia ossificans progressiva (FOP), resulting in the transformation of soft tissue into bone. This unexpected link points toward the importance of developmental biology processes in tumorigenesis and provides an extensive experience in mechanistic understanding and drug development hard-won by FOP researchers to pediatric neurooncology. Here, we review the literature in both fields and identify potential areas for collaboration and rapid advancement for patients of both diseases.


Subject(s)
Activin Receptors, Type I/genetics , Brain Stem Neoplasms/genetics , Glioma/genetics , Mutation/genetics , Myositis Ossificans/genetics , Animals , Brain Stem Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Glioma/pathology , Humans , Myositis Ossificans/pathology
18.
Nat Genet ; 46(5): 457-461, 2014 May.
Article in English | MEDLINE | ID: mdl-24705252

ABSTRACT

Diffuse intrinsic pontine gliomas (DIPGs) are highly infiltrative malignant glial neoplasms of the ventral pons that, due to their location within the brain, are unsuitable for surgical resection and consequently have a universally dismal clinical outcome. The median survival time is 9-12 months, with neither chemotherapeutic nor targeted agents showing substantial survival benefit in clinical trials in children with these tumors. We report the identification of recurrent activating mutations in the ACVR1 gene, which encodes a type I activin receptor serine/threonine kinase, in 21% of DIPG samples. Strikingly, these somatic mutations (encoding p.Arg206His, p.Arg258Gly, p.Gly328Glu, p.Gly328Val, p.Gly328Trp and p.Gly356Asp substitutions) have not been reported previously in cancer but are identical to mutations found in the germ line of individuals with the congenital childhood developmental disorder fibrodysplasia ossificans progressiva (FOP) and have been shown to constitutively activate the BMP-TGF-ß signaling pathway. These mutations represent new targets for therapeutic intervention in this otherwise incurable disease.


Subject(s)
Activin Receptors, Type I/genetics , Brain Stem Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Glioma/genetics , Mutation, Missense/genetics , Base Sequence , Brain Stem Neoplasms/classification , Child , Cohort Studies , Exome/genetics , Glioma/classification , Humans , Molecular Sequence Data , Myositis Ossificans/genetics , Sequence Analysis, DNA , Signal Transduction/genetics
19.
Nat Genet ; 46(5): 451-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24705254

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a fatal brain cancer that arises in the brainstem of children, with no effective treatment and near 100% fatality. The failure of most therapies can be attributed to the delicate location of these tumors and to the selection of therapies on the basis of assumptions that DIPGs are molecularly similar to adult disease. Recent studies have unraveled the unique genetic makeup of this brain cancer, with nearly 80% found to harbor a p.Lys27Met histone H3.3 or p.Lys27Met histone H3.1 alteration. However, DIPGs are still thought of as one disease, with limited understanding of the genetic drivers of these tumors. To understand what drives DIPGs, we integrated whole-genome sequencing with methylation, expression and copy number profiling, discovering that DIPGs comprise three molecularly distinct subgroups (H3-K27M, silent and MYCN) and uncovering a new recurrent activating mutation affecting the activin receptor gene ACVR1 in 20% of DIPGs. Mutations in ACVR1 were constitutively activating, leading to SMAD phosphorylation and increased expression of the downstream activin signaling targets ID1 and ID2. Our results highlight distinct molecular subgroups and novel therapeutic targets for this incurable pediatric cancer.


Subject(s)
Activin Receptors, Type I/genetics , Brain Stem Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Glioma/genetics , Animals , Brain Stem Neoplasms/classification , Child , DNA Copy Number Variations , DNA Methylation , Gene Expression Profiling , Glioma/classification , Humans , Inhibitor of Differentiation Protein 1/metabolism , Inhibitor of Differentiation Protein 2/metabolism , Phosphorylation , Sequence Analysis, DNA , Smad Proteins/metabolism , Zebrafish
20.
Clin Cancer Res ; 19(21): 5940-51, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23918606

ABSTRACT

PURPOSE: To provide rationale for using phosphoinositide 3-kinase (PI3K) and/or mitogen-activated protein kinase (MAPK) pathway inhibitors to treat rhabdomyosarcomas, a major cause of pediatric and adolescent cancer deaths. EXPERIMENTAL DESIGN: The prevalence of PI3K/MAPK pathway activation in rhabdomyosarcoma clinical samples was assessed using immunohistochemistry. Compensatory signaling and cross-talk between PI3K/MAPK pathways was determined in rhabdomyosarcoma cell lines following p110α short hairpin RNA-mediated depletion. Pharmacologic inhibition of reprogrammed signaling in stable p110α knockdown lines was used to determine the target-inhibition profile inducing maximal growth inhibition. The in vitro and in vivo efficacy of inhibitors of TORC1/2 (AZD8055), MEK (AZD6244), and P13K/mTOR (NVP-BEZ235) was evaluated alone and in pairwise combinations. RESULTS: PI3K pathway activation was seen in 82.5% rhabdomyosarcomas with coactivated MAPK in 36% and 46% of alveolar and embryonal subtypes, respectively. p110α knockdown in cell lines over the short and long term was associated with compensatory expression of other p110 isoforms, activation of the MAPK pathway, and cross-talk to reactivate the PI3K pathway. Combinations of PI3K pathway and MAP-ERK kinase (MEK) inhibitors synergistically inhibited cell growth in vitro. Treatment of RD cells with AZD8055 plus AZD6244 blocked reciprocal pathway activation, as evidenced by reduced AKT/ERK/S6 phosphorylation. In vivo, the synergistic effect on growth and changes in pharmacodynamic biomarkers was recapitulated using the AZD8055/AZD6244 combination but not NVP-BEZ235/AZD6244. Pharmacokinetic analysis provided evidence of drug-drug interaction with both combinations. CONCLUSIONS: Dual PI3K/MAPK pathway activation and compensatory signaling in both rhabdomyosarcoma subtypes predict a lack of clinical efficacy for single agents targeting either pathway, supporting a therapeutic strategy combining a TORC1/2 with a MEK inhibitor.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Drug Synergism , Enzyme Activation/drug effects , Female , Gene Knockdown Techniques , Humans , Morpholines/administration & dosage , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Rhabdomyosarcoma/drug therapy , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...