Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 2123, 2024 01 24.
Article in English | MEDLINE | ID: mdl-38267500

ABSTRACT

Quiescence, a reversible state of cell-cycle arrest, is an important state during both normal development and cancer progression. For example, in glioblastoma (GBM) quiescent glioblastoma stem cells (GSCs) play an important role in re-establishing the tumour, leading to relapse. While most studies have focused on identifying differentially expressed genes between proliferative and quiescent cells as potential drivers of this transition, recent studies have shown the importance of protein oscillations in controlling the exit from quiescence of neural stem cells. Here, we have undertaken a genome-wide bioinformatic inference approach to identify genes whose expression oscillates and which may be good candidates for controlling the transition to and from the quiescent cell state in GBM. Our analysis identified, among others, a list of important transcription regulators as potential oscillators, including the stemness gene SOX2, which we verified to oscillate in quiescent GSCs. These findings expand on the way we think about gene regulation and introduce new candidate genes as key regulators of quiescence.


Subject(s)
Glioblastoma , Neural Stem Cells , Humans , Glioblastoma/genetics , Cell Division , Computational Biology , Gene Expression , SOXB1 Transcription Factors/genetics
2.
J Neurooncol ; 161(3): 451-467, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36757526

ABSTRACT

INTRODUCTION: Brain tumors cause morbidity and mortality in part through peritumoral brain edema. The current main treatment for peritumoral brain edema are corticosteroids. Due to the increased recognition of their side-effect profile, there is growing interest in finding alternatives to steroids but there is little formal study of animal models of peritumoral brain edema. This study aims to summarize the available literature. METHODS: A systematic search was undertaken of 5 literature databases (Medline, Embase, CINAHL, PubMed and the Cochrane Library). The generic strategy was to search for various terms associated with "brain tumors", "brain edema" and "animal models". RESULTS: We identified 603 reports, of which 112 were identified as relevant for full text analysis that studied 114 peritumoral brain edema animal models. We found significant heterogeneity in the species and strain of tumor-bearing animals, tumor implantation method and edema assessment. Most models did not produce appreciable brain edema and did not test for observable manifestations thereof. CONCLUSION: No animal model currently exists that enable the investigation of novel candidates for the treatment of peritumoral brain edema. With current interest in alternative treatments for peritumoral brain edema, there is an unmet need for clinically relevant animal models.


Subject(s)
Brain Edema , Brain Neoplasms , Animals , Humans , Magnetic Resonance Imaging/methods , Brain Neoplasms/pathology , Edema/complications , Brain Edema/complications , Brain/pathology
3.
Drug Resist Updat ; 65: 100888, 2022 12.
Article in English | MEDLINE | ID: mdl-36332495

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide, with non-small cell lung cancer (NSCLC) being the predominant histological subtype. Despite the emergence of targeted and immune-based therapies that have considerably improved the clinical outcomes of selected patients, the overall NSCLC survival rate remains poor. NSCLC patients experience clinical relapse mainly because of chemoresistance. One promising therapeutic approach is targeting specific molecular vulnerabilities that are associated with the metabolic reprogramming of cancer cells. This strategy relies on evidence that cancer cells rewire their metabolism to sustain their uncontrolled growth as well as invasive and metastatic properties, promoting adaptive resistance to chemo-radiotherapy. A critical component of this malignant transformation is the increased dependency on high levels of heat shock proteins (HSPs), which support the elevated protein folding demand and quality control of misfolded oncoproteins. Here, we provide an overview of the literature on metabolism reprogramming, deregulation of mitochondrion and on the role of HSPs in promoting malignancy in lung and other cancer types. A particular focus is dedicated to the role of mitochondrial HSP60 (HSPD1) in NSCLC metabolism and drug resistance for the potential development of new resistance-defying anti-HSP drugs.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mitochondria , Drug Resistance
4.
Neuro Oncol ; 24(4): 541-553, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34543427

ABSTRACT

BACKGROUND: Microtubes (MTs), cytoplasmic extensions of glioma cells, are important cell communication structures promoting invasion and treatment resistance through network formation. MTs are abundant in chemoresistant gliomas, in particular, glioblastomas (GBMs), while they are uncommon in chemosensitive IDH-mutant and 1p/19q co-deleted oligodendrogliomas. The aim of this study was to identify potential signaling pathways involved in MT formation. METHODS: Bioinformatics analysis of TCGA was performed to analyze differences between GBM and oligodendroglioma. Patient-derived GBM stem cell lines were used to investigate MT formation under transforming growth factor-beta (TGF-ß) stimulation and inhibition in vitro and in vivo in an orthotopic xenograft model. RNA sequencing and proteomics were performed to detect commonalities and differences between GBM cell lines stimulated with TGF-ß. RESULTS: Analysis of TCGA data showed that the TGF-ß pathway is highly activated in GBMs compared to oligodendroglial tumors. We demonstrated that TGF-ß1 stimulation of GBM cell lines promotes enhanced MT formation and communication via calcium signaling. Inhibition of the TGF-ß pathway significantly reduced MT formation and its associated invasion in vitro and in vivo. Downstream of TGF-ß, we identified thrombospondin 1 (TSP1) as a potential mediator of MT formation in GBM through SMAD activation. TSP1 was upregulated upon TGF-ß stimulation and enhanced MT formation, which was inhibited by TSP1 shRNAs in vitro and in vivo. CONCLUSION: TGF-ß and its downstream mediator TSP1 are important mediators of the MT network in GBM and blocking this pathway could potentially help to break the complex MT-driven invasion/resistance network.


Subject(s)
Glioblastoma , Glioma , Oligodendroglioma , Glioblastoma/pathology , Humans , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
5.
J Exp Clin Cancer Res ; 40(1): 248, 2021 Aug 07.
Article in English | MEDLINE | ID: mdl-34364401

ABSTRACT

BACKGROUND: The identification of novel targets is of paramount importance to develop more effective drugs and improve the treatment of non-small cell lung cancer (NSCLC), the leading cause of cancer-related deaths worldwide. Since cells alter their metabolic rewiring during tumorigenesis and along cancer progression, targeting key metabolic players and metabolism-associated proteins represents a valuable approach with a high therapeutic potential. Metabolic fitness relies on the functionality of heat shock proteins (HSPs), molecular chaperones that facilitate the correct folding of metabolism enzymes and their assembly in macromolecular structures. METHODS: Gene fitness was determined by bioinformatics analysis from available datasets from genetic screenings. HSPD1 expression was evaluated by immunohistochemistry from formalin-fixed paraffin-embedded tissues from NSCLC patients. Real-time proliferation assays with and without cytotoxicity reagents, colony formation assays and cell cycle analyses were used to monitor growth and drug sensitivity of different NSCLC cells in vitro. In vivo growth was monitored with subcutaneous injections in immune-deficient mice. Cell metabolic activity was analyzed through extracellular metabolic flux analysis. Specific knockouts were introduced by CRISPR/Cas9. RESULTS: We show heat shock protein family D member 1 (HSPD1 or HSP60) as a survival gene ubiquitously expressed in NSCLC and associated with poor patients' prognosis. HSPD1 knockdown or its chemical disruption by the small molecule KHS101 induces a drastic breakdown of oxidative phosphorylation, and suppresses cell proliferation both in vitro and in vivo. By combining drug profiling with transcriptomics and through a whole-genome CRISPR/Cas9 screen, we demonstrate that HSPD1-targeted anti-cancer effects are dependent on oxidative phosphorylation and validated molecular determinants of KHS101 sensitivity, in particular, the creatine-transporter SLC6A8 and the subunit of the cytochrome c oxidase complex COX5B. CONCLUSIONS: These results highlight mitochondrial metabolism as an attractive target and HSPD1 as a potential theranostic marker for developing therapies to combat NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Chaperonin 60/metabolism , Lung Neoplasms/genetics , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Disease Models, Animal , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , Survival Analysis
6.
Nat Commun ; 12(1): 3898, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34162854

ABSTRACT

One topical area of supramolecular chemistry is the binding of anionic species but despite the importance of anions in diverse cellular processes and for cancer development, anion receptors or 'binders' have received little attention as potential anti-cancer therapeutics. Here we report self-assembling trimetallic cryptands (e.g. [L2(Metal)3]6+ where Metal = Cu2+, Zn2+ or Mn2+) which can encapsulate a range of anions and which show metal-dependent differences in chemical and biological reactivities. In cell studies, both [L2Cu3]6+ and [L2Zn3]6+ complexes are highly toxic to a range of human cancer cell lines and they show significant metal-dependent selective activity towards cancer cells compared to healthy, non-cancerous cells (by up to 2000-fold). The addition of different anions to the complexes (e.g. PO43-, SO42- or PhOPO32-) further alters activity and selectivity allowing the activity to be modulated via a self-assembly process. The activity is attributed to the ability to either bind or hydrolyse phosphate esters and mechanistic studies show differential and selective inhibition of multiple kinases by both [L2Cu3]6+ and [L2Zn3]6+ complexes but via different mechanisms.


Subject(s)
Anions/chemistry , Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Metals/chemistry , A549 Cells , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/pharmacology , Crystallography, X-Ray , HCT116 Cells , HT29 Cells , Humans , Inhibitory Concentration 50 , Neoplasms/metabolism , Neoplasms/pathology , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/metabolism
7.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33707311

ABSTRACT

Patients with glioblastoma (GBM) have a poor prognosis, and inefficient delivery of drugs to tumors represents a major therapeutic hurdle. Hematopoietic stem cell (HSC)-derived myeloid cells efficiently home to GBM and constitute up to 50% of intratumoral cells, making them highly appropriate therapeutic delivery vehicles. Because myeloid cells are ubiquitously present in the body, we recently established a lentiviral vector containing matrix metalloproteinase 14 (MMP14) promoter, which is active specifically in tumor-infiltrating myeloid cells as opposed to myeloid cells in other tissues, and resulted in a specific delivery of transgenes to brain metastases in HSC gene therapy. Here, we used this novel approach to target transforming growth factor beta (TGFß) as a key tumor-promoting factor in GBM. Transplantation of HSCs transduced with lentiviral vector expressing green fluorescent protein (GFP) into lethally irradiated recipient mice was followed by intracranial implantation of GBM cells. Tumor-infiltrating HSC progeny was characterized by flow cytometry. In therapy studies, mice were transplanted with HSCs transduced with lentiviral vector expressing soluble TGFß receptor II-Fc fusion protein under MMP14 promoter. This TGFß-blocking therapy was compared with the targeted tumor irradiation, the combination of the two therapies, and control. Tumor growth and survival were quantified (statistical significance determined by t-test and log-rank test). T cell memory response was probed through a repeated tumor challenge. Myeloid cells were the most abundant HSC-derived population infiltrating GBM. TGFß-blocking HSC gene therapy in combination with irradiation significantly reduced tumor burden as compared with monotherapies and the control, and significantly prolonged survival as compared with the control and TGFß-blocking monotherapy. Long-term protection from GBM was achieved only with the combination treatment (25% of the mice) and was accompanied by a significant increase in CD8+ T cells at the tumor implantation site following tumor rechallenge. We demonstrated a preclinical proof-of-principle for tumor myeloid cell-specific HSC gene therapy in GBM. In the clinic, HSC gene therapy is being successfully used in non-cancerous brain disorders and the feasibility of HSC gene therapy in patients with glioma has been demonstrated in the context of bone marrow protection. This indicates an opportunity for clinical translation of our therapeutic approach.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy , Glioblastoma/therapy , Hematopoietic Stem Cell Transplantation , Immunoglobulin Fc Fragments/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Immunoglobulin Fc Fragments/metabolism , Matrix Metalloproteinase 14/genetics , Mice, Inbred C57BL , Promoter Regions, Genetic , Proof of Concept Study , Radiotherapy, Adjuvant , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Burden
8.
iScience ; 24(1): 102022, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33506187

ABSTRACT

Chaperonins play an important role in folding newly synthesized or translocated proteins in all organisms. The bacterial chaperonin GroEL has served as a model system for the understanding of these proteins. In comparison, its human homolog, known as mitochondrial heat shock protein family member D1 (HSPD1) is poorly understood. Here, we present the structure of HSPD1 in the apo state determined by cryo-electron microscopy (cryo-EM). Unlike GroEL, HSPD1 forms mostly single ring assemblies in the absence of co-chaperonin (HSPE1). Comparison with GroEL shows a rotation and increased flexibility of the apical domain. Together with published structures of the HSPD1/HSPE1 co-chaperonin complex, this work gives insight into the structural changes that occur during the catalytic cycle. This new understanding of HSPD1 structure and its rearrangements upon complex formation may provide new insights for the development of HSPD1-targeting treatments against a diverse range of diseases including glioblastoma.

9.
J Colloid Interface Sci ; 584: 669-683, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33223243

ABSTRACT

Porous silica nanoparticles (PSiNPs) have long attracted interest in drug delivery research. However, conventional synthesis methods for sub-100 nm, functionalised PSiNPs typically give poor monodispersity, reproducibility, or involve complex synthetic protocols. We report a facile, reproducible, and cost-effective one-pot method for the synthesis of cancer targeting and pH responsive PSiNPs in this size range, without the need for post-synthetic modification. This was achieved by using monodisperse l-arginine (Arg)/ poly(acrylic acid) (PAA) polyelectrolyte complexes (PECs) as soft templates for silane hydrolysis and condensation. Highly uniform PSiNPs with tunable size control between 42 and 178 nm and disordered pore structure (1.1-2.7 nm) were obtained. Both PAA and Arg were retained within the PSiNPs, which enabled a high doxorubicin hydrochloride (Dox) loading capacity (22% w/w) and a 4-fold increase in drug release under weakly acidic pH compared to physiological pH. The surface presentation of Arg conferred significantly higher intracellular accumulation of Arg/PAA-PSiNPs in patient-derived glioblastoma cells compared to non-tumorigenic neural progenitor cells, which effectively translated to lower IC50 values for Dox-loaded Arg/PAA-PSiNPs than non-functionalised PSiNPs. This work brings forward new insights for the development of monodisperse PSiNPs with highly desirable built-in functionalities for biomedical applications.


Subject(s)
Nanoparticles , Neoplasms , Pharmaceutical Preparations , Doxorubicin/pharmacology , Drug Carriers , Drug Delivery Systems , Humans , Hydrogen-Ion Concentration , Polyelectrolytes , Porosity , Reproducibility of Results , Silicon Dioxide
10.
Mol Genet Genomic Med ; 8(10): e1429, 2020 10.
Article in English | MEDLINE | ID: mdl-32720471

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.


Subject(s)
Cell Death/drug effects , Cerebellar Neoplasms/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Medulloblastoma/metabolism , Repressor Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cerebellum/metabolism , Humans , Repressor Proteins/genetics
11.
J Cell Sci ; 132(19)2019 10 10.
Article in English | MEDLINE | ID: mdl-31515278

ABSTRACT

Tumor stem cells and malignant multicellular networks have been separately implicated in the therapeutic resistance of glioblastoma multiforme (GBM), the most aggressive type of brain cancer in adults. Here, we show that small-molecule inhibition of RHO-associated serine/threonine kinase proteins (ROCKi) significantly promoted the outgrowth of neurite-like cell projections in cultures of heterogeneous patient-derived GBM stem-like cells. These projections formed de novo-induced cellular network (iNet) 'webs', which regressed after withdrawal of ROCKi. Connected cells within the iNet web exhibited long range Ca2+ signal transmission, and significant lysosomal and mitochondrial trafficking. In contrast to their less-connected vehicle control counterparts, iNet cells remained viable and proliferative after high-dose radiation. These findings demonstrate a link between ROCKi-regulated cell projection dynamics and the formation of radiation-resistant multicellular networks. Our study identifies means to reversibly induce iNet webs ex vivo, and may thereby accelerate future studies into the biology of GBM cellular networks.


Subject(s)
Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Neurites/metabolism , Calcium Signaling/physiology , Cell Line, Tumor , Cell Movement/physiology , Humans , Immunoblotting , Lysosomes/metabolism , Mitochondria/metabolism , Neuronal Outgrowth/physiology , Phenotype , Protein Serine-Threonine Kinases/metabolism
12.
Proc Natl Acad Sci U S A ; 116(13): 6435-6440, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30846550

ABSTRACT

Glioblastoma multiforme (GBM; grade IV astrocytoma) is the most prevalent and aggressive form of primary brain cancer. A subpopulation of multipotent cells termed GBM cancer stem cells (CSCs) play a critical role in tumor initiation, tumor maintenance, metastasis, drug resistance, and recurrence following surgery. Here we report the identification of a small molecule, termed RIPGBM, from a cell-based chemical screen that selectively induces apoptosis in multiple primary patient-derived GBM CSC cultures. The cell type-dependent selectivity of this compound appears to arise at least in part from redox-dependent formation of a proapoptotic derivative, termed cRIPGBM, in GBM CSCs. cRIPGBM induces caspase 1-dependent apoptosis by binding to receptor-interacting protein kinase 2 (RIPK2) and acting as a molecular switch, which reduces the formation of a prosurvival RIPK2/TAK1 complex and increases the formation of a proapoptotic RIPK2/caspase 1 complex. In an orthotopic intracranial GBM CSC tumor xenograft mouse model, RIPGBM was found to significantly suppress tumor formation in vivo. Our chemical genetics-based approach has identified a drug candidate and a potential drug target that provide an approach to the development of treatments for this devastating disease.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Animals , Astrocytes , Cell Line, Tumor , Disease Models, Animal , Drug Delivery Systems , Drug Evaluation, Preclinical , Female , Glioblastoma , Heterografts , High-Throughput Screening Assays , Humans , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Pyroptosis/drug effects , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
13.
J Pathol ; 247(4): 422-434, 2019 04.
Article in English | MEDLINE | ID: mdl-30565681

ABSTRACT

The overall survival for patients with primary glioblastoma is very poor. Glioblastoma contains a subpopulation of glioma stem cells (GSC) that are responsible for tumour initiation, treatment resistance and recurrence. PPARα is a transcription factor involved in the control of lipid, carbohydrate and amino acid metabolism. We have recently shown that PPARα gene and protein expression is increased in glioblastoma and has independent clinical prognostic significance in multivariate analyses. In this work, we report that PPARα is overexpressed in GSC compared to foetal neural stem cells. To investigate the role of PPARα in GSC, we knocked down its expression using lentiviral transduction with short hairpin RNA (shRNA). Transduced GSC were tagged with luciferase and stereotactically xenografted into the striatum of NOD-SCID mice. Bioluminescent and magnetic resonance imaging showed that knockdown (KD) of PPARα reduced the tumourigenicity of GSC in vivo. PPARα-expressing control GSC xenografts formed invasive histological phenocopies of human glioblastoma, whereas PPARα KD GSC xenografts failed to establish viable intracranial tumours. PPARα KD GSC showed significantly reduced proliferative capacity and clonogenic potential in vitro with an increase in cellular senescence. In addition, PPARα KD resulted in significant downregulation of the stem cell factors c-Myc, nestin and SOX2. This was accompanied by downregulation of the PPARα-target genes and key regulators of fatty acid oxygenation ACOX1 and CPT1A, with no compensatory increase in glycolytic flux. These data establish the aberrant overexpression of PPARα in GSC and demonstrate that this expression functions as an important regulator of tumourigenesis, linking self-renewal and the malignant phenotype in this aggressive cancer stem cell subpopulation. We conclude that targeting GSC PPARα expression may be a therapeutically beneficial strategy with translational potential as an adjuvant treatment. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , PPAR alpha/metabolism , RNA, Small Interfering/pharmacology , Animals , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques/methods , Humans , Lentivirus , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , Phenotype , Signal Transduction/physiology , Transplantation, Heterologous , Tumor Cells, Cultured
14.
Sci Transl Med ; 10(454)2018 08 15.
Article in English | MEDLINE | ID: mdl-30111643

ABSTRACT

Pharmacological inhibition of uncontrolled cell growth with small-molecule inhibitors is a potential strategy for treating glioblastoma multiforme (GBM), the most malignant primary brain cancer. We showed that the synthetic small-molecule KHS101 promoted tumor cell death in diverse GBM cell models, independent of their tumor subtype, and without affecting the viability of noncancerous brain cell lines. KHS101 exerted cytotoxic effects by disrupting the mitochondrial chaperone heat shock protein family D member 1 (HSPD1). In GBM cells, KHS101 promoted aggregation of proteins regulating mitochondrial integrity and energy metabolism. Mitochondrial bioenergetic capacity and glycolytic activity were selectively impaired in KHS101-treated GBM cells. In two intracranial patient-derived xenograft tumor models in mice, systemic administration of KHS101 reduced tumor growth and increased survival without discernible side effects. These findings suggest that targeting of HSPD1-dependent metabolic pathways might be an effective strategy for treating GBM.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Energy Metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Thiazoles/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Chaperonin 60/metabolism , Citric Acid Cycle/drug effects , Disease Models, Animal , Energy Metabolism/drug effects , Glioblastoma/genetics , Glycolysis/drug effects , Humans , Metabolic Networks and Pathways/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neoplasm Invasiveness , Stress, Physiological/drug effects , Survival Analysis , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
15.
J Control Release ; 281: 29-41, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29753957

ABSTRACT

Magnetic Resonance Image-guided Focused Ultrasound (MRgFUS) has been used to achieve transient blood brain barrier (BBB) opening without tissue injury. Delivery of a targeted ultrasonic wave causes an interaction between administered microbubbles and the capillary bed resulting in enhanced vessel permeability. The use of MRgFUS in the brainstem has not previously been shown but could provide value in the treatment of tumours such as Diffuse Intrinsic Pontine Glioma (DIPG) where the intact BBB has contributed to the limited success of chemotherapy. Our primary objective was to determine whether the use of MRgFUS in this eloquent brain region could be performed without histological injury and functional deficits. Our secondary objective was to select an effective chemotherapeutic against patient derived DIPG cell lines and demonstrate enhanced brainstem delivery when combined with MRgFUS in vivo. Female Sprague Dawley rats were randomised to one of four groups: 1) Microbubble administration but no MRgFUS treatment; 2) MRgFUS only; 3) MRgFUS + microbubbles; and 4) MRgFUS + microbubbles + cisplatin. Physiological assessment was performed by monitoring of heart and respiratory rates. Motor function and co-ordination were evaluated by Rotarod and grip strength testing. Histological analysis for haemorrhage (H&E), neuronal nuclei (NeuN) and apoptosis (cleaved Caspase-3) was also performed. A drug screen of eight chemotherapy agents was conducted in three patient-derived DIPG cell lines (SU-DIPG IV, SU-DIPG XIII and SU-DIPG XVII). Doxorubicin was identified as an effective agent. NOD/SCID/GAMMA (NSG) mice were subsequently administered with 5 mg/kg of intravenous doxorubicin at the time of one of the following: 1) Microbubbles but no MRgFUS; 2) MRgFUS only; 3) MRgFUS + microbubbles and 4) no intervention. Brain specimens were extracted at 2 h and doxorubicin quantification was conducted using liquid chromatography mass spectrometry (LC/MS). BBB opening was confirmed by contrast enhancement on T1-weighted MR imaging and positive Evans blue staining of the brainstem. Normal cardiorespiratory parameters were preserved. Grip strength and Rotarod testing demonstrating no decline in performance across all groups. Histological analysis showed no evidence of haemorrhage, neuronal loss or increased apoptosis. Doxorubicin demonstrated cytotoxicity against all three cell lines and is known to have poor BBB permeability. Quantities measured in the brainstem of NSG mice were highest in the group receiving MRgFUS and microbubbles (431.5 ng/g). This was significantly higher than in mice who received no intervention (7.6 ng/g). Our data demonstrates both the preservation of histological and functional integrity of the brainstem following MRgFUS for BBB opening and the ability to significantly enhance drug delivery to the region, giving promise to the treatment of brainstem-specific conditions.


Subject(s)
Antineoplastic Agents/administration & dosage , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Doxorubicin/administration & dosage , Glioma/drug therapy , Ultrasonic Waves , Animals , Antineoplastic Agents/therapeutic use , Brain/metabolism , Brain Stem , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Doxorubicin/therapeutic use , Drug Carriers , Drug Liberation , Female , Mice, SCID , Microbubbles , Permeability , Rats, Sprague-Dawley , Tissue Distribution
16.
SLAS Discov ; 23(8): 862-868, 2018 09.
Article in English | MEDLINE | ID: mdl-29543559

ABSTRACT

Organoid methodology provides a platform for the ex vivo investigation of the cellular and molecular mechanisms underlying brain development and disease. The high-grade brain tumor glioblastoma multiforme (GBM) is considered a cancer of unmet clinical need, in part due to GBM cell infiltration into healthy brain parenchyma, making complete surgical resection improbable. Modeling the process of GBM invasion in real time is challenging as it requires both tumor and neural tissue compartments. Here, we demonstrate that human GBM spheroids possess the ability to spontaneously infiltrate early-stage cerebral organoids (eCOs). The resulting formation of hybrid organoids demonstrated an invasive tumor phenotype that was distinct from noncancerous adult neural progenitor (NP) spheroid incorporation into eCOs. These findings provide a basis for the modeling and quantification of the GBM infiltration process using a stem-cell-based organoid approach, and may be used for the identification of anti-GBM invasion strategies.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Biomarkers , Brain Neoplasms/metabolism , Cell Culture Techniques , Cell Movement , Fluorescent Antibody Technique , Glioblastoma/metabolism , Humans , Immunohistochemistry , Neoplasm Invasiveness , Neoplasm Staging , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Organoids
17.
Sci Transl Med ; 10(422)2018 01 03.
Article in English | MEDLINE | ID: mdl-29298869

ABSTRACT

Immune checkpoint inhibitors, including those targeting programmed cell death protein 1 (PD-1), are reshaping cancer therapeutic strategies. Evidence suggests, however, that tumor response and patient survival are determined by tumor programmed death ligand 1 (PD-L1) expression. We hypothesized that preconditioning of the tumor immune microenvironment using targeted, virus-mediated interferon (IFN) stimulation would up-regulate tumor PD-L1 protein expression and increase cytotoxic T cell infiltration, improving the efficacy of subsequent checkpoint blockade. Oncolytic viruses (OVs) represent a promising form of cancer immunotherapy. For brain tumors, almost all studies to date have used direct intralesional injection of OV, because of the largely untested belief that intravenous administration will not deliver virus to this site. We show, in a window-of-opportunity clinical study, that intravenous infusion of oncolytic human Orthoreovirus (referred to herein as reovirus) leads to infection of tumor cells subsequently resected as part of standard clinical care, both in high-grade glioma and in brain metastases, and increases cytotoxic T cell tumor infiltration relative to patients not treated with virus. We further show that reovirus up-regulates IFN-regulated gene expression, as well as the PD-1/PD-L1 axis in tumors, via an IFN-mediated mechanism. Finally, we show that addition of PD-1 blockade to reovirus enhances systemic therapy in a preclinical glioma model. These results support the development of combined systemic immunovirotherapy strategies for the treatment of both primary and secondary tumors in the brain.


Subject(s)
Brain Neoplasms/therapy , Oncolytic Viruses/pathogenicity , Animals , Glioma/therapy , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism
18.
Stem Cell Reports ; 8(1): 125-139, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28076755

ABSTRACT

Patients with glioblastoma die from local relapse despite surgery and high-dose radiotherapy. Resistance to radiotherapy is thought to be due to efficient DNA double-strand break (DSB) repair in stem-like cells able to survive DNA damage and repopulate the tumor. We used clinical samples and patient-derived glioblastoma stem cells (GSCs) to confirm that the DSB repair protein RAD51 is highly expressed in GSCs, which are reliant on RAD51-dependent DSB repair after radiation. RAD51 expression and RAD51 foci numbers fall when these cells move toward astrocytic differentiation. In GSCs, the small-molecule RAD51 inhibitors RI-1 and B02 prevent RAD51 focus formation, reduce DNA DSB repair, and cause significant radiosensitization. We further demonstrate that treatment with these agents combined with radiation promotes loss of stem cells defined by SOX2 expression. This indicates that RAD51-dependent repair represents an effective and specific target in GSCs.


Subject(s)
DNA Repair , Glioma/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Rad51 Recombinase/genetics , Radiation Tolerance/genetics , Animals , Cell Differentiation , Cell Line, Tumor , DNA Damage/radiation effects , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Glioma/pathology , Humans , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/metabolism , Radiation-Sensitizing Agents/pharmacology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Xenograft Model Antitumor Assays
19.
Oncotarget ; 7(27): 41473-41487, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27203741

ABSTRACT

In contrast to primary tumors, the understanding of macrophages within metastases is very limited. In order to compare macrophage phenotypes between different metastatic sites, we established a pre-clinical mouse model of intracranial breast cancer metastasis in which cancer lesions develop simultaneously within the brain parenchyma and the dura. This mimics a situation that is commonly occurring in the clinic. Flow cytometry analysis revealed significant differences in the activation state of metastasis-associated macrophages (MAMs) at the two locations. Concurrently, gene expression analysis identified significant differences in molecular profiles of cancer cells that have metastasized to the brain parenchyma as compared to the dura. This included differences in inflammation-related pathways, NF-kB1 activity and cytokine profiles. The most significantly upregulated cytokine in brain parenchyma- versus dura-derived cancer cells was Lymphotoxin ß and a gain-of-function approach demonstrated a direct involvement of this factor in the M2 polarization of parenchymal MAMs. This established a link between metastatic site-specific properties of cancer cells and the MAM activation state.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Cell Movement , Cell Polarity , Macrophages/physiology , Animals , Brain/pathology , Cell Line, Tumor , Encephalitis/pathology , Female , Humans , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Tumor Microenvironment/immunology
20.
Oncoscience ; 2(3): 252-62, 2015.
Article in English | MEDLINE | ID: mdl-25897422

ABSTRACT

MicroRNA expression can be exploited to define tumor prognosis and stratification for precision medicine. It remains unclear whether prognostic microRNA signatures are exclusively tumor grade and/or molecular subtype-specific, or whether common signatures of aggressive clinical behavior can be identified. Here, we defined microRNAs that are associated with good and poor prognosis in grade III and IV gliomas using data from The Cancer Genome Atlas. Pathway analysis of microRNA targets that are differentially expressed in good and poor prognosis glioma identified a link to oligodendrocyte development. Notably, a microRNA expression profile that is characteristic of a specific oligodendrocyte precursor cell type (OP1) correlates with microRNA expression from 597 of these tumors and is consistently associated with poor patient outcome in grade III and IV gliomas. Our study reveals grade-independent and subtype-independent prognostic molecular signatures in high-grade glioma and provides a framework for investigating the mechanisms of brain tumor aggressiveness.

SELECTION OF CITATIONS
SEARCH DETAIL
...