Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4616, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816355

ABSTRACT

Dynamic regulation of gene expression is fundamental for cellular adaptation to exogenous stressors. P-TEFb-mediated pause-release of RNA polymerase II (Pol II) is a conserved regulatory mechanism for synchronous transcriptional induction in response to heat shock, but this pro-survival role has not been examined in the applied context of cancer therapy. Using model systems of pediatric high-grade glioma, we show that rapid genome-wide reorganization of active chromatin facilitates P-TEFb-mediated nascent transcriptional induction within hours of exposure to therapeutic ionizing radiation. Concurrent inhibition of P-TEFb disrupts this chromatin reorganization and blunts transcriptional induction, abrogating key adaptive programs such as DNA damage repair and cell cycle regulation. This combination demonstrates a potent, synergistic therapeutic potential agnostic of glioma subtype, leading to a marked induction of tumor cell apoptosis and prolongation of xenograft survival. These studies reveal a central role for P-TEFb underpinning the early adaptive response to radiotherapy, opening avenues for combinatorial treatment in these lethal malignancies.


Subject(s)
Gene Expression Regulation, Neoplastic , Glioma , Positive Transcriptional Elongation Factor B , Humans , Glioma/radiotherapy , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Animals , Positive Transcriptional Elongation Factor B/metabolism , Positive Transcriptional Elongation Factor B/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/radiation effects , Mice , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Transcription, Genetic/radiation effects , Apoptosis/radiation effects , Apoptosis/genetics , Brain Neoplasms/radiotherapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , DNA Repair/radiation effects , Xenograft Model Antitumor Assays
3.
bioRxiv ; 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38116029

ABSTRACT

Polycomb Repressive Complex 2 (PRC2)-mediated histone H3K27 tri-methylation (H3K27me3) recruits canonical PRC1 (cPRC1) to maintain heterochromatin. In early development, polycomb-regulated genes are connected through long-range 3D interactions which resolve upon differentiation. Here, we report that polycomb looping is controlled by H3K27me3 spreading and regulates target gene silencing and cell fate specification. Using glioma-derived H3 Lys-27-Met (H3K27M) mutations as tools to restrict H3K27me3 deposition, we show that H3K27me3 confinement concentrates the chromatin pool of cPRC1, resulting in heightened 3D interactions mirroring chromatin architecture of pluripotency, and stringent gene repression that maintains cells in progenitor states to facilitate tumor development. Conversely, H3K27me3 spread in pluripotent stem cells, following neural differentiation or loss of the H3K36 methyltransferase NSD1, dilutes cPRC1 concentration and dissolves polycomb loops. These results identify the regulatory principles and disease implications of polycomb looping and nominate histone modification-guided distribution of reader complexes as an important mechanism for nuclear compartment organization. Highlights: The confinement of H3K27me3 at PRC2 nucleation sites without its spreading correlates with increased 3D chromatin interactions.The H3K27M oncohistone concentrates canonical PRC1 that anchors chromatin loop interactions in gliomas, silencing developmental programs.Stem and progenitor cells require factors promoting H3K27me3 confinement, including H3K36me2, to maintain cPRC1 loop architecture.The cPRC1-H3K27me3 interaction is a targetable driver of aberrant self-renewal in tumor cells.

4.
bioRxiv ; 2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36747867

ABSTRACT

Dynamic regulation of gene expression is fundamental for cellular adaptation to exogenous stressors. PTEFb-mediated pause-release of RNA polymerase II (Pol II) is a conserved regulatory mechanism for synchronous transcriptional induction in response to heat shock, but this pro-survival role has not been examined in the applied context of cancer therapy. Using model systems of pediatric high-grade glioma, we show that rapid genome-wide reorganization of active chromatin facilitates PTEFb-mediated nascent transcriptional induction within hours of exposure to therapeutic ionizing radiation. Concurrent inhibition of PTEFb disrupts this chromatin reorganization and blunts transcriptional induction, abrogating key adaptive programs such as DNA damage repair and cell cycle regulation. This combination demonstrates a potent, synergistic therapeutic potential agnostic of glioma subtype, leading to a marked induction of tumor cell apoptosis and prolongation of xenograft survival. These studies reveal a central role for PTEFb underpinning the early adaptive response to radiotherapy, opening new avenues for combinatorial treatment in these lethal malignancies.

5.
Pediatr Blood Cancer ; 70(3): e30190, 2023 03.
Article in English | MEDLINE | ID: mdl-36602034

ABSTRACT

The event-free survival of pediatric low-grade gliomas is poor, and patients often require multiple treatment strategies. While MEK and RAF inhibitors are efficacious in early-phase trials, not all patients respond, and many experience progression following completion of therapy. Evaluating combination therapies that may enhance efficacy or prolong disease stabilization is warranted. We report our institutional experience using concurrent trametinib and lenalidomide in the treatment of primary pediatric central and peripheral nervous system tumors. Two of four patients using this combination therapy experienced severe thromboembolic events, necessitating discontinuation of therapy. This combination requires further investigation, and we urge caution if used.


Subject(s)
Pyridones , Pyrimidinones , Humans , Child , Lenalidomide/adverse effects , Pyridones/adverse effects , Pyrimidinones/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Proto-Oncogene Proteins B-raf
7.
Clin Cancer Res ; 28(11): 2409-2424, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35344040

ABSTRACT

PURPOSE: Tumor relapse after radiotherapy is a major hurdle in treating pediatric H3K27M-mutant diffuse midline gliomas (DMG). Radiotherapy-induced stress increases association of BCL2 family of proteins with BH3 pro-apoptotic activators preventing apoptosis. We hypothesized that inhibition of radiotherapy-induced BCL2 with a clinically relevant inhibitor, venetoclax, will block BCL2 activity leading to increased apoptosis. BCL2 has never been implicated in DMG as a radiotherapy-induced resistant mechanism. EXPERIMENTAL DESIGN: We performed an integrated genomic analysis to determine genes responsible for radioresistance and a targeted drug screen to identify drugs that synergize with radiation in DMG. Effect of venetoclax on radiation-naïve and 6 Gy radiation on cells was evaluated by studying cell death, changes in BCL2 phosphorylation, reactive oxygen species (ROS), and apoptosis, as well as BCL2 association with BH3 apoptosis initiators. The efficacy of combining venetoclax with radiation was evaluated in vivo using orthotopic xenograft models. RESULTS: BCL2 was identified as a key regulator of tumor growth after radiation in DMGs. Radiation sensitizes DMGs to venetoclax treatment independent of p53 status. Venetoclax as a monotherapy was not cytotoxic to DMG cells. Postradiation venetoclax treatment significantly increased cell death, reduced BCL2-BIM association, and augmented mitochondrial ROS leading to increased apoptosis. Combining venetoclax with radiotherapy significantly enhanced the survival of mice with DMG tumors. CONCLUSIONS: This study shows that venetoclax impedes the antiapoptotic function of radiation-induced BCL2 in DMG, leading to increased apoptosis. Results from these preclinical studies demonstrate the potential use of the BCL2 inhibitor venetoclax combined with radiotherapy for pediatric DMG.


Subject(s)
Antineoplastic Agents , Glioma , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cell Line, Tumor , Glioma/drug therapy , Glioma/genetics , Glioma/radiotherapy , Humans , Mice , Neoplasm Recurrence, Local/drug therapy , Proto-Oncogene Proteins c-bcl-2 , Radiation, Ionizing , Reactive Oxygen Species , Sulfonamides
9.
J Neuropathol Exp Neurol ; 80(4): 345-353, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33749791

ABSTRACT

Diffuse midline gliomas (DMGs) are incurable pediatric tumors with extraordinarily limited treatment options. Decades of clinical trials combining conventional chemotherapies with radiation therapy have failed to improve these outcomes, demonstrating the need to identify and validate druggable biologic targets within this disease. NTRK1/2/3 fusions are found in a broad range of pediatric cancers, including high-grade gliomas and a subset of DMGs. Phase 1/2 studies of TRK inhibitors have demonstrated good tolerability, effective CNS penetration, and promising objective responses across all patients with TRK fusion-positive cancers, but their use has not been explored in TRK fusion-positive DMG. Here, we report 3 cases of NTRK fusions co-occurring within H3K27M-positive pontine diffuse midline gliomas. We employ a combination of single-cell and bulk transcriptome sequencing from TRK fusion-positive DMG to describe the phenotypic consequences of this co-occurring alteration. We then use ex vivo short-culture assays to evaluate the potential response to TRK inhibition in this disease. Together, these data highlight the importance of routine molecular characterization of these highly aggressive tumors and identify a small subset of patients that may benefit from currently available targeted therapies.


Subject(s)
Brain Stem Neoplasms/genetics , Glioma/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Membrane Glycoproteins/genetics , Mutation/genetics , Receptor, trkB/genetics , Receptor, trkC/genetics , Brain Stem Neoplasms/diagnostic imaging , Child , Child, Preschool , Fatal Outcome , Female , Glioma/diagnostic imaging , Humans , Male
11.
Cell Rep ; 31(1): 107485, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32268092

ABSTRACT

Histone 3 gene mutations are the eponymous drivers in diffuse midline gliomas (DMGs), aggressive pediatric brain cancers for which no curative therapy currently exists. These recurrent oncohistones induce a global loss of repressive H3K27me3 residues and broad epigenetic dysregulation. In order to identify therapeutically targetable dependencies within this disease context, we performed an RNAi screen targeting epigenetic/chromatin-associated genes in patient-derived DMG cultures. This identified AFF4, the scaffold protein of the super elongation complex (SEC), as a molecular dependency in DMG. Interrogation of SEC function demonstrates a key role for maintaining clonogenic potential while promoting self-renewal of tumor stem cells. Small-molecule inhibition of SEC using clinically relevant CDK9 inhibitors restores regulatory RNA polymerase II pausing, promotes cellular differentiation, and leads to potent anti-tumor effect both in vitro and in patient-derived xenograft models. These studies present a rationale for further exploration of SEC inhibition as a promising therapeutic approach to this intractable disease.


Subject(s)
Glioma/genetics , Histones/genetics , Transcriptional Elongation Factors/metabolism , Brain/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Chromatin/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Gene Expression Regulation, Neoplastic/genetics , Glioma/metabolism , Histones/metabolism , Humans , Transcriptional Elongation Factors/genetics
12.
Childs Nerv Syst ; 35(11): 2043-2046, 2019 11.
Article in English | MEDLINE | ID: mdl-31367784

ABSTRACT

PURPOSE: Radiation-induced injury is a well-described toxicity in children receiving radiation therapy for tumors of the central nervous system. Standard therapy has historically consisted primarily of high-dose corticosteroids, which carry significant side effects. Preclinical models suggest that radiation necrosis may be mediated in part through vascular endothelial growth factor (VEGF) overexpression, providing the rationale for use of VEGF inhibitors in the treatment of CNS radiation necrosis. We present the first prospective experience examining the safety, feasibility, neurologic outcomes, and imaging characteristics of bevacizumab therapy for CNS radiation necrosis in children. METHODS: Seven patients between 1 and 25 years of age with neurologic deterioration and MRI findings consistent with radiation injury or necrosis were enrolled on an IRB-approved pilot feasibility study. Patients received bevacizumab at a dose of 10 mg/kg intravenously every 2 weeks for up to 6 total doses. RESULTS: Five patients (83%) were able to wean off corticosteroid therapy during the study period and 4 patients (57%) demonstrated improvement in serial neurologic exams. All patients demonstrated a decrease in T1-weighted post-gadolinium enhancement on MRI, while 5 (71%) showed a decrease in FLAIR signal. Four patients developed a progressive disease of their underlying tumor during bevacizumab therapy. CONCLUSIONS: Our experience lends support to the safety and feasibility of bevacizumab administration for the treatment of radiation necrosis for appropriately selected patients within the pediatric population.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Brain Diseases/drug therapy , Central Nervous System Neoplasms/radiotherapy , Radiation Injuries/drug therapy , Radiotherapy/adverse effects , Adolescent , Brain Diseases/diagnostic imaging , Brain Diseases/etiology , Brain Diseases/physiopathology , Child , Child, Preschool , Dexamethasone/therapeutic use , Dose Fractionation, Radiation , Feasibility Studies , Female , Glucocorticoids/therapeutic use , Humans , Magnetic Resonance Imaging , Male , Necrosis , Pilot Projects , Radiation Dose Hypofractionation , Radiation Injuries/diagnostic imaging , Radiation Injuries/etiology , Radiation Injuries/physiopathology
13.
Fam Cancer ; 18(3): 327-330, 2019 07.
Article in English | MEDLINE | ID: mdl-30919136

ABSTRACT

Familial adenomatous polyposis (FAP) is a cancer predisposition syndrome driven by germline loss-of-function of the APC gene and phenotypically manifests with intestinal polyposis and a variety of extra-intestinal bone and soft tissue tumors. Craniopharyngioma is not a well-described FAP-associated tumor, however, six cases have been reported in adults, all demonstrating ectopic location and adamantinomatous histology. We report the first case of craniopharyngioma associated with FAP in a pediatric patient. A seven-year-old girl who presented with headache and vomiting was found on magnetic resonance imaging to have a suprasellar mass with cystic extension to the pre-pontine space. The tumor represented an adamantinomatous craniopharyngioma (aCP) with nuclear ß-catenin expression. Whole exome sequencing confirmed a CTNNB1 activating point mutation and a germline APC frameshift variant. This case represents the first FAP-associated craniopharyngioma in childhood…. expanding our understanding of the molecular underpinnings driving tumorigenesis in this unique patient.


Subject(s)
Adenomatous Polyposis Coli/complications , Craniopharyngioma/complications , Pituitary Neoplasms/complications , beta Catenin/genetics , Adenomatous Polyposis Coli/genetics , Child , Craniopharyngioma/diagnostic imaging , Craniopharyngioma/genetics , Female , Genes, APC , Germ-Line Mutation , Humans , Phenotype , Pituitary Neoplasms/diagnostic imaging , Pituitary Neoplasms/genetics , Point Mutation , beta Catenin/metabolism
14.
J Neuropathol Exp Neurol ; 76(6): 418-423, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28498973

ABSTRACT

Chordomas are rare bony neoplasms usually unassociated with a familial tumor predisposition syndrome. The peak incidence of this midline axial skeletal tumor is in adulthood but when very young children are affected, consideration should be given to occurrence within the tuberous sclerosis (TS) complex, especially when presenting in neonates <3 months of age. To call attention to this association, we present a brachyury-immunopositive chordoma occurring in the skull base of a 2-month-old male infant who was later realized to have metastases to the subcutaneous tissues and lungs, as well as rhabdomyoma of the heart and renal cysts/angiomyolipomas, that is, characteristic features of the TS complex. We review the limited literature on this topic.


Subject(s)
Chordoma/pathology , Skull Base Neoplasms/pathology , Tuberous Sclerosis/pathology , Angiomyolipoma/diagnostic imaging , Angiomyolipoma/pathology , Chordoma/diagnostic imaging , Chordoma/genetics , Heart Neoplasms/diagnostic imaging , Heart Neoplasms/secondary , Humans , Infant , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/secondary , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/secondary , Magnetic Resonance Imaging , Male , Neoplasm Metastasis , Rhabdomyoma/pathology , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/pathology , Skin Neoplasms/secondary , Skull Base Neoplasms/diagnostic imaging , Skull Base Neoplasms/genetics , Tuberous Sclerosis/diagnostic imaging , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/genetics
15.
J Pediatr Hematol Oncol ; 38(5): e154-7, 2016 07.
Article in English | MEDLINE | ID: mdl-26840078

ABSTRACT

The proportion and clinical characteristics of Gardner fibromas (GAFs) that are sporadic versus familial adenomatous polyposis (FAP)-associated have not been clearly established. We report on 7 patients diagnosed with GAF who underwent APC sequencing and duplication/deletion testing. Three (43%) were found to have underlying APC germline perturbations consistent with FAP; these patients had multifocal (1) or large; unresectable (2) GAFs. The 4 patients with negative APC testing each had a single resectable GAF. ß-catenin reactivity was noted in all FAP-associated GAFs and in 1/4 APC wild-type cases. FAP-associated GAFs may be less common than sporadic GAFs and can demonstrate clinically distinct features.


Subject(s)
Fibroma , Gardner Syndrome , Genes, APC , Germ-Line Mutation , Adenomatous Polyposis Coli/pathology , Child, Preschool , Female , Humans , Infant , Male , Retrospective Studies , Sequence Analysis, DNA , beta Catenin/metabolism
16.
Pediatr Blood Cancer ; 63(3): 551-3, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26485542

ABSTRACT

Monosomy 7 is a well-documented cytogenetic aberration in pediatric acute myeloid leukemia (AML) and may occur in combinations with molecular abnormalities including PTPN11 mutation. PTPN11 mutations contribute to leukemogenesis through upregulation of Ras pathway signaling. We present the case of a 3-year-old female with AML with monosomy 7 and somatic PTPN11 mutation who was refractory to conventional AML chemotherapy but responded to a novel regimen of azacitidine and sorafenib followed by stem cell transplantation. Combination therapy with azacitidine and sorafenib may be an effective therapeutic strategy for patients with AML with Ras pathway abnormalities.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Agents/administration & dosage , Azacitidine/administration & dosage , Chromosome Deletion , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Child, Preschool , Chromosomes, Human, Pair 7 , Drug Therapy, Combination , Female , Humans , Mutation , Niacinamide/administration & dosage , Sorafenib , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...