Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters








Publication year range
1.
Genome Biol ; 25(1): 264, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39390467

ABSTRACT

BACKGROUND: Diffuse invasion of glioblastoma cells through normal brain tissue is a key contributor to tumor aggressiveness, resistance to conventional therapies, and dismal prognosis in patients. A deeper understanding of how components of the tumor microenvironment (TME) contribute to overall tumor organization and to programs of invasion may reveal opportunities for improved therapeutic strategies. RESULTS: Towards this goal, we apply a novel computational workflow to a spatiotemporally profiled GBM xenograft cohort, leveraging the ability to distinguish human tumor from mouse TME to overcome previous limitations in the analysis of diffuse invasion. Our analytic approach, based on unsupervised deconvolution, performs reference-free discovery of cell types and cell activities within the complete GBM ecosystem. We present a comprehensive catalogue of 15 tumor cell programs set within the spatiotemporal context of 90 mouse brain and TME cell types, cell activities, and anatomic structures. Distinct tumor programs related to invasion align with routes of perivascular, white matter, and parenchymal invasion. Furthermore, sub-modules of genes serving as program network hubs are highly prognostic in GBM patients. CONCLUSION: The compendium of programs presented here provides a basis for rational targeting of tumor and/or TME components. We anticipate that our approach will facilitate an ecosystem-level understanding of the immediate and long-term consequences of such perturbations, including the identification of compensatory programs that will inform improved combinatorial therapies.


Subject(s)
Brain Neoplasms , Glioblastoma , Neoplasm Invasiveness , Tumor Microenvironment , Glioblastoma/pathology , Glioblastoma/genetics , Animals , Humans , Mice , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Spatio-Temporal Analysis
2.
Nat Commun ; 14(1): 3062, 2023 05 27.
Article in English | MEDLINE | ID: mdl-37244935

ABSTRACT

Self-renewal is a crucial property of glioblastoma cells that is enabled by the choreographed functions of chromatin regulators and transcription factors. Identifying targetable epigenetic mechanisms of self-renewal could therefore represent an important step toward developing effective treatments for this universally lethal cancer. Here we uncover an epigenetic axis of self-renewal mediated by the histone variant macroH2A2. With omics and functional assays deploying patient-derived in vitro and in vivo models, we show that macroH2A2 shapes chromatin accessibility at enhancer elements to antagonize transcriptional programs of self-renewal. macroH2A2 also sensitizes cells to small molecule-mediated cell death via activation of a viral mimicry response. Consistent with these results, our analyses of clinical cohorts indicate that high transcriptional levels of this histone variant are associated with better prognosis of high-grade glioma patients. Our results reveal a targetable epigenetic mechanism of self-renewal controlled by macroH2A2 and suggest additional treatment approaches for glioblastoma patients.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Histones/genetics , Histones/metabolism , Glioblastoma/metabolism , Gene Expression Regulation, Neoplastic , Chromatin/metabolism , Epigenesis, Genetic , Cell Line, Tumor , Neoplastic Stem Cells/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism
3.
Neurosurg Focus ; 50(2): E9, 2021 02.
Article in English | MEDLINE | ID: mdl-33524950

ABSTRACT

OBJECTIVE: The aim of this study was to demonstrate the in vivo safety and antitumor effect of a novel recombinant vesicular stomatitis virus (VSV): G protein less (GLESS)-fusion-associated small transmembrane (FAST)-VSV. METHODS: Viral infection efficiency and cell proliferation were detected using an inverted fluorescence microscope and alarmaBlue assay, respectively. To evaluate the safety of the virus, different doses of GLESS-FAST-VSV and a positive control virus (VSV∆M51) were injected into normal F344 rats and C57BL/6 mice, and each animal's weight, survival time, and pathological changes were examined on the following day. To evaluate the efficacy of the virus, RG2 and GL261 cells were used to construct rat and mouse glioma models, respectively, via a stereotactic method. After multiple intratumoral injections of the virus, tumor growth (size) and the survival time of the animals were observed. RESULTS: In vitro experiments showed that GLESS-FAST-VSV could infect and kill brain tumor cells and had less toxic effects on normal cells. After direct injection of GLESS-FAST-VSV into the animal brains, all animals tolerated the virus well, and no animal death, encephalitis, or ventriculitis was observed. In contrast, all animals that received brain injections of VSV∆M51 in the brain died. Moreover, multiple injections of GLESS-FAST-VSV in brain tumors significantly prolonged the survival of normal-immunity animals harboring brain tumors. CONCLUSIONS: GLESS-FAST-VSV exhibited little neurotoxicity and could be injected directly into the tumor to effectively inhibit tumor growth and prolong the survival of normal-immunity animals, laying a theoretical foundation for the early application of such viruses in clinical trials.


Subject(s)
Glioma , Vesicular Stomatitis , Animals , Glioma/drug therapy , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred F344 , Vesicular stomatitis Indiana virus
4.
Cancer Gene Ther ; 28(7-8): 739-744, 2021 08.
Article in English | MEDLINE | ID: mdl-32678303

ABSTRACT

Central nervous system (CNS) atypical teratoid/rhabdoid tumor (AT/RT) is a rare, aggressive tumor that most often affects very young children. The common decisive molecular defect in AT/RT has been shown to be a single genetic alteration, i.e., the loss of hSNF5 gene that encodes for a subunit of the SWI/SNF complex that modulates chromatin remodeling activities. As a result, AT/RT cells display unregulated cell proliferation due to the dysfunction of an important epigenetic control. We have previously demonstrated the preclinical efficacy of the oncolytic double-deleted vaccinia virus (VVDD) against AT/RT. Here we report the establishment of a modified VVDD engineered to express wild type hSNF5 gene. We show that this reconstructed vaccinia virus retains comparable infectivity and in vitro cytotoxicity of the parent strain. However, in addition, hSNF5-arming of VVDD results in a decreased cell cycle S phase population and down-regulation of cyclin D1. These findings suggest that hSNF5-arming of VVDD may increase the efficacy in the treatment of AT/RT and validates, as a proof-of-concept, an experimental approach to enhance the effective use of novel modified oncolytic viruses in the treatment of tumors with loss of a tumor suppressor gene function.


Subject(s)
Central Nervous System/pathology , Oncolytic Viruses/metabolism , Rhabdoid Tumor/genetics , Rhabdoid Tumor/virology , SMARCB1 Protein/metabolism , Vaccinia virus/metabolism , Animals , Disease Models, Animal , Female , Humans , Mice , Xenograft Model Antitumor Assays
5.
Nat Commun ; 11(1): 4997, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020472

ABSTRACT

Despite a deeper molecular understanding, human glioblastoma remains one of the most treatment refractory and fatal cancers. It is known that the presence of macrophages and microglia impact glioblastoma tumorigenesis and prevent durable response. Herein we identify the dual function cytokine IL-33 as an orchestrator of the glioblastoma microenvironment that contributes to tumorigenesis. We find that IL-33 expression in a large subset of human glioma specimens and murine models correlates with increased tumor-associated macrophages/monocytes/microglia. In addition, nuclear and secreted functions of IL-33 regulate chemokines that collectively recruit and activate circulating and resident innate immune cells creating a pro-tumorigenic environment. Conversely, loss of nuclear IL-33 cripples recruitment, dramatically suppresses glioma growth, and increases survival. Our data supports the paradigm that recruitment and activation of immune cells, when instructed appropriately, offer a therapeutic strategy that switches the focus from the cancer cell alone to one that includes the normal host environment.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Interleukin-33/metabolism , Animals , Brain Neoplasms/mortality , Carcinogenesis , Cell Nucleus/metabolism , Cytokines/metabolism , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Glioma/mortality , Humans , Inflammation , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, SCID , Microglia , Survival Analysis , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Microenvironment/immunology
6.
Biomaterials ; 252: 120105, 2020 09.
Article in English | MEDLINE | ID: mdl-32417652

ABSTRACT

Despite extensive molecular characterization, human glioblastoma remains a fatal disease with survival rates measured in months. Little improvement is seen with standard surgery, radiotherapy and chemotherapy. Clinical progress is hampered by the inability to detect and target glioblastoma disease reservoirs based on a diffuse invasive pattern and the presence of molecular and phenotypic heterogeneity. The goal of this study was to target the invasive and stem-like glioblastoma cells that evade first-line treatments using agents capable of delivering imaging enhancers or biotherapeutic cargo. To accomplish this, a combinatorial phage display library was biopanned against glioblastoma cell model systems that accurately recapitulate the intra- and inter-tumor heterogeneity and infiltrative nature of the disease. Candidate peptides were screened for specificity and ability to target glioblastoma cells in vivo. Cargo-conjugated peptides delivered contrast-enhancing agents to highly infiltrative tumor populations in intracranial xenograft models without the obvious need for blood brain barrier disruption. Simultaneous use of five independent targeting peptides provided greater coverage of this complex tumor and selected peptides have the capacity to deliver a therapeutic cargo (oncolytic virus VSVΔM51) to the tumor cells in vivo. Herein, we have identified a series of peptides with utility as an innovative platform to assist in targeting glioblastoma for the purpose of diagnostic or prognostic imaging, image-guided surgery, and/or improved delivery of therapeutic agents to glioblastoma cells implicated in disease relapse.


Subject(s)
Brain Neoplasms , Glioblastoma , Oncolytic Viruses , Animals , Cell Line, Tumor , Glioblastoma/drug therapy , Humans , Peptides
7.
Proc Natl Acad Sci U S A ; 116(38): 19098-19108, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31471491

ABSTRACT

Glioblastoma multiforme (GBM) is the most deadly brain tumor, and currently lacks effective treatment options. Brain tumor-initiating cells (BTICs) and orthotopic xenografts are widely used in investigating GBM biology and new therapies for this aggressive disease. However, the genomic characteristics and molecular resemblance of these models to GBM tumors remain undetermined. We used massively parallel sequencing technology to decode the genomes and transcriptomes of BTICs and xenografts and their matched tumors in order to delineate the potential impacts of the distinct growth environments. Using data generated from whole-genome sequencing of 201 samples and RNA sequencing of 118 samples, we show that BTICs and xenografts resemble their parental tumor at the genomic level but differ at the mRNA expression and epigenomic levels, likely due to the different growth environment for each sample type. These findings suggest that a comprehensive genomic understanding of in vitro and in vivo GBM model systems is crucial for interpreting data from drug screens, and can help control for biases introduced by cell-culture conditions and the microenvironment in mouse models. We also found that lack of MGMT expression in pretreated GBM is linked to hypermutation, which in turn contributes to increased genomic heterogeneity and requires new strategies for GBM treatment.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genomics/methods , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Tumor Microenvironment/genetics , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Brain Neoplasms/genetics , Case-Control Studies , Cell Proliferation , DNA Methylation , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Glioblastoma/genetics , Humans , Male , Mice , Mice, SCID , Middle Aged , Neoplastic Stem Cells/metabolism , Transcriptome , Tumor Cells, Cultured , Whole Genome Sequencing , Xenograft Model Antitumor Assays
8.
PLoS One ; 13(8): e0202860, 2018.
Article in English | MEDLINE | ID: mdl-30153289

ABSTRACT

BACKGROUND: Temozolomide (TMZ) is active against glioblastomas (GBM) in which the O6-methylguanine-DNA methyltransferase (MGMT) gene is silenced. However, even in responsive cases, its beneficial effect is undermined by the emergence of drug resistance. Here, we tested whether inhibition of poly (ADP-ribose) polymerase-1 and -2 (PARP) enhanced the effectiveness of TMZ. METHODS: Using patient derived brain tumor initiating cells (BTICs) and orthotopic xenografts as models of newly diagnosed and recurrent high-grade glioma, we assessed the effects of TMZ, ABT-888, and the combination of TMZ and ABT-888 on the viability of BTICs and survival of tumor-bearing mice. We also studied DNA damage repair, checkpoint protein phosphorylation, and DNA replication in mismatch repair (MMR) deficient cells treated with TMZ and TMZ plus ABT-888. RESULTS: Cells and xenografts derived from newly diagnosed MGMT methylated high-grade gliomas were sensitive to TMZ while those derived from unmethylated and recurrent gliomas were typically resistant. ABT-888 had no effect on the viability of BTICs or tumor bearing mice, but co-treatment with TMZ restored sensitivity in resistant cells and xenografts from newly diagnosed unmethylated gliomas and recurrent gliomas with MSH6 mutations. In contrast, the addition of ABT-888 to TMZ had little sensitizing effect on cells and xenografts derived from newly diagnosed methylated gliomas. In a model of acquired TMZ resistance mediated by loss of MMR gene MSH6, re-sensitization to TMZ by ABT-888 was accompanied by persistent DNA strand breaks, re-engagement of checkpoint kinase signaling, and interruption of DNA synthesis. CONCLUSION: In laboratory models, the addition of ABT-888 to TMZ overcame resistance to TMZ.


Subject(s)
Benzimidazoles/pharmacology , Drug Resistance, Neoplasm/drug effects , Glioma/pathology , Temozolomide/pharmacology , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Gene Knockdown Techniques , Humans , Mice , Neoplasm Grading , RNA, Small Interfering/genetics
10.
Biomed Pharmacother ; 95: 1201-1208, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28931212

ABSTRACT

BACKGROUND: Malignant glioma still has a poor prognosis and remains incurable. Although temozolomide (TMZ) has demonstrated antitumor activity, its use recently has been halted because of some patients' resistance to this drug. New treatments are desperately needed. An oncolytic virus (virotherapy) is being developed as a novel cancer therapy. We have previously reported that recombinant Vesicular Stomatitis Virus (VSV-ΔM51) and double deleted Vaccinia Virus (vvDD) infected and killed glioma cell lines in vitro and prolonged survival in animal glioma models. As a proposed ex vivo test, the oncolytic potential of VSV-ΔM51 and vvDD in the established human brain tumor stem cells (BTSCs) and the differentiated cells from fresh brain tumor tissues in vitro were further investigated. METHODS: BTSCs from fresh surgical glioblastoma multiforme (GBM) specimens were isolated and cultured, and the characterization of BTSCs were tested. The sensitivity of BTSCs to TMZ and the susceptibility of TMZ resistant BTSCs and their differentiated cells to both oncolytic viruses were examined. RESULTS: The BTSC spheres cultured had all the characteristics of stem cells. The GFP-labeled VSV-ΔM51 and vvDD could infect TMZ resistant BTSCs and cause cytopathic effects. The VSV-ΔM51and vvDD inhibited the self-renewal activity of TMZ resistant BTSCs. And the VSV-ΔM51and vvDD also infected and caused cytopathic effects in differentiated BTSCs. CONCLUSION: VSV-ΔM51and vvDD could infect and kill both the TMZ resistant BTSCs and the differentiated compartments of GBMs in vitro, suggesting that they may be an effective treatment supplement for GBM therapy, particularly for TMZ resistant GBM patients.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm , Neoplastic Stem Cells/pathology , Recombination, Genetic/genetics , Vaccinia virus/genetics , Vesicular Stomatitis/genetics , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Temozolomide , Virus Replication
11.
Nat Commun ; 82017 02 15.
Article in English | MEDLINE | ID: mdl-28198370

ABSTRACT

Small-molecule inhibitor of apoptosis (IAP) antagonists, called Smac mimetic compounds (SMCs), sensitize tumours to TNF-α-induced killing while simultaneously blocking TNF-α growth-promoting activities. SMCs also regulate several immunomodulatory properties within immune cells. We report that SMCs synergize with innate immune stimulants and immune checkpoint inhibitor biologics to produce durable cures in mouse models of glioblastoma in which single agent therapy is ineffective. The complementation of activities between these classes of therapeutics is dependent on cytotoxic T-cell activity and is associated with a reduction in immunosuppressive T-cells. Notably, the synergistic effect is dependent on type I IFN and TNF-α signalling. Furthermore, our results implicate an important role for TNF-α-producing cytotoxic T-cells in mediating the anti-cancer effects of immune checkpoint inhibitors when combined with SMCs. Overall, this combinatorial approach could be highly effective in clinical application as it allows for cooperative and complimentary mechanisms in the immune cell-mediated death of cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Interferon-alpha/immunology , Interferon-beta/immunology , Thiazoles/pharmacology , Adaptive Immunity/drug effects , Animals , Antineoplastic Agents/chemical synthesis , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Cell Line, Tumor , Female , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/immunology , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/mortality , Humans , Immunity, Innate/drug effects , Immunologic Memory , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/immunology , Interferon-alpha/genetics , Interferon-alpha/pharmacology , Interferon-beta/genetics , Interferon-beta/pharmacology , Mice , Poly I-C/pharmacology , Signal Transduction , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Thiazoles/chemical synthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology , Vesiculovirus/genetics , Vesiculovirus/immunology , Xenograft Model Antitumor Assays
12.
Oncotarget ; 7(37): 59360-59376, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27449082

ABSTRACT

Glioblastoma (GBM) is the most lethal and aggressive adult brain tumor, requiring the development of efficacious therapeutics. Towards this goal, we screened five genetically distinct patient-derived brain-tumor initiating cell lines (BTIC) with a unique collection of small molecule epigenetic modulators from the Structural Genomics Consortium (SGC). We identified multiple hits that inhibited the growth of BTICs in vitro, and further evaluated the therapeutic potential of EZH2 and HDAC inhibitors due to the high relevance of these targets for GBM. We found that the novel SAM-competitive EZH2 inhibitor UNC1999 exhibited low micromolar cytotoxicity in vitro on a diverse collection of BTIC lines, synergized with dexamethasone (DEX) and suppressed tumor growth in vivo in combination with DEX. In addition, a unique brain-penetrant class I HDAC inhibitor exhibited cytotoxicity in vitro on a panel of BTIC lines and extended survival in combination with TMZ in an orthotopic BTIC model in vivo. Finally, a combination of EZH2 and HDAC inhibitors demonstrated synergy in vitro by augmenting apoptosis and increasing DNA damage. Our findings identify key epigenetic modulators in GBM that regulate BTIC growth and survival and highlight promising combination therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Drug Screening Assays, Antitumor/methods , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Glioblastoma/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Pyridones/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Dexamethasone/therapeutic use , Drug Synergism , Drug Therapy, Combination , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Mice, SCID , Molecular Targeted Therapy , Pyridones/pharmacology , Small Molecule Libraries , Xenograft Model Antitumor Assays
13.
Neuro Oncol ; 18(8): 1088-1098, 2016 08.
Article in English | MEDLINE | ID: mdl-26962017

ABSTRACT

BACKGROUND: Myxoma virus (MYXV) is a promising oncolytic agent and is highly effective against immortalized glioma cells but less effective against brain tumor initiating cells (BTICs), which are believed to mediate glioma development/recurrence. MYXV encodes various proteins to attenuate host cell apoptosis, including an antiapoptotic Bcl-2 homologue known as M011L. Such proteins may limit the ability of MYXV to kill BTICs, which have heightened resistance to apoptosis. We hypothesized that infecting BTICs with an M011L-deficient MYXV construct would overcome BTIC resistance to MYXV. METHODS: We used patient-derived BTICs to evaluate the efficacy of M011L knockout virus (vMyx-M011L-KO) versus wild-type MYXV (vMyx-WT) and characterized the mechanism of virus-induced cell death in vitro. To extend our findings in a novel immunocompetent animal model, we derived, cultured, and characterized a C57Bl/6J murine BTIC (mBTIC0309) from a spontaneous murine glioma and evaluated vMyx-M011L-KO efficacy with and without temozolomide (TMZ) in mBTIC0309-bearing mice. RESULTS: We demonstrated that vMyx-M011L-KO induces apoptosis in BTICs, dramatically increasing sensitivity to the virus. vMyx-WT failed to induce apoptosis as M011L protein prevented Bax activation and cytochrome c release. In vivo, intracranial implantation of mBTIC0309 generated tumors that closely recapitulated the pathological and molecular profile of human gliomas. Treatment of tumor-bearing mice with vMyx-M011L-KO significantly prolonged survival in immunocompetent-but not immunodeficient-mouse models, an effect that is significantly enhanced in combination with TMZ. CONCLUSIONS: Our data suggest that vMyx-M011L-KO is an effective, well-tolerated, proapoptotic oncolytic virus and a strong candidate for clinical translation.

14.
Clin Cancer Res ; 22(15): 3860-75, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27006494

ABSTRACT

PURPOSE: Glioblastoma is one of the most lethal cancers in humans, and with existing therapy, survival remains at 14.6 months. Current barriers to successful treatment include their infiltrative behavior, extensive tumor heterogeneity, and the presence of a stem-like population of cells, termed brain tumor-initiating cells (BTIC) that confer resistance to conventional therapies. EXPERIMENTAL DESIGN: To develop therapeutic strategies that target BTICs, we focused on a repurposing approach that explored already-marketed (clinically approved) drugs for therapeutic potential against patient-derived BTICs that encompass the genetic and phenotypic heterogeneity of glioblastoma observed clinically. RESULTS: Using a high-throughput in vitro drug screen, we found that montelukast, clioquinol, and disulfiram (DSF) were cytotoxic against a large panel of patient-derived BTICs. Of these compounds, disulfiram, an off-patent drug previously used to treat alcoholism, in the presence of a copper supplement, showed low nanomolar efficacy in BTICs including those resistant to temozolomide and the highly infiltrative quiescent stem-like population. Low dose DSF-Cu significantly augmented temozolomide activity in vitro, and importantly, prolonged in vivo survival in patient-derived BTIC models established from both newly diagnosed and recurrent tumors. Moreover, we found that in addition to acting as a potent proteasome inhibitor, DSF-Cu functionally impairs DNA repair pathways and enhances the effects of DNA alkylating agents and radiation. These observations suggest that DSF-Cu inhibits proteasome activity and augments the therapeutic effects of DNA-damaging agents (temozolomide and radiation). CONCLUSIONS: DSF-Cu should be considered as an adjuvant therapy for the treatment of patients with glioblastoma in both newly diagnosed and recurrent settings. Clin Cancer Res; 22(15); 3860-75. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Copper/pharmacology , Dacarbazine/analogs & derivatives , Disulfiram/pharmacology , Glioblastoma/metabolism , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Repair , Dacarbazine/pharmacology , Disease Models, Animal , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , Female , Gene Expression Profiling , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , High-Throughput Screening Assays , Humans , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Proteasome Endopeptidase Complex/metabolism , Temozolomide , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Neuro Oncol ; 17(8): 1086-94, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25605818

ABSTRACT

BACKGROUND: Brain tumor-initiating cells (BTICs) are stem-like cells hypothesized to form a disease reservoir that mediates tumor recurrence in high-grade gliomas. Oncolytic virotherapy uses replication-competent viruses to target and kill malignant cells and has been evaluated in clinic for glioma therapy with limited results. Myxoma virus (MyxV) is a safe and highly effective oncolytic virus (OV) in conventional glioma models but, as seen with other OVs, is only modestly effective for patient-derived BTICs. The objective of this study was to determine whether MyxV treatment against human BTICs could be improved by combining chemotherapeutics and virotherapy. METHODS: A 73-compound library of drug candidates in clinical use or preclinical development was screened to identify compounds that sensitize human BTICs to MyxV treatment in vitro, and synergy was evaluated mathematically in lead compounds using Chou-Talalay analyses. The effects of combination therapy on viral gene expression and viral replication were also assessed. RESULTS: Eleven compounds that enhance MyxV efficacy were identified, and 6 were shown to synergize with the virus using Chou-Talalay analyses. Four of the synergistic compounds were shown to significantly increase viral gene expression, indicating a potential mechanism for synergy. Three highly synergistic compounds (axitinib, a VEGFR inhibitor; rofecoxib, a cyclooxygenase-2 inhibitor; and pemetrexed, a folate anti-metabolite) belong to classes of compounds that have not been previously shown to synergize with oncolytic viruses in vitro. CONCLUSIONS: This study has identified multiple novel drug candidates that synergistically improve MyxV efficacy in a preclinical BTIC glioma model.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/virology , Oncolytic Virotherapy , Antineoplastic Agents/administration & dosage , Axitinib , Brain Neoplasms/virology , Cell Line, Tumor , Combined Modality Therapy , Glioblastoma/virology , Humans , Imidazoles/administration & dosage , Imidazoles/therapeutic use , In Vitro Techniques , Indazoles/administration & dosage , Indazoles/therapeutic use , Myxoma virus/genetics , Myxoma virus/physiology , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Small Molecule Libraries
16.
Neuro Oncol ; 17(6): 822-31, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25395461

ABSTRACT

BACKGROUND: Neurocutaneous melanocytosis (NCM) is a rare congenital disorder that presents with pigmented cell lesions of the brain or leptomeninges in children with large or multiple congenital melanocytic nevi. Although the exact pathological processes involved are currently unclear, NCM appears to arise from an abnormal development of melanoblasts or melanocyte precursors. Currently, it has an extremely poor prognosis due to rapid disease progression and lack of effective treatment modalities. METHODS: In this study, we report on an experimental approach to examining NCM cells by establishing subcutaneous tumors in nude mice, which can be further expanded for conducting molecular and drug sensitivity experiments. RESULTS: Analysis of the NRAS gene-coding sequences of an established NCM cell line (YP-MEL) and NCM patient cells revealed heterogeneity in NRAS Q61K that activated mutation and possibly consequential differential sensitivity to MEK inhibition. Gene expression studies were performed to compare the molecular profiles of NCM cells with normal skin fibroblasts. In vitro cytotoxicity screens of libraries of targeted small-molecule inhibitors revealed prospective agents for further evaluation. CONCLUSIONS: Our studies provide an experimental platform for the generation of NCM cells for preclinical studies and the production of molecular and in vitro data with which to identify druggable targets for the treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Melanosis/drug therapy , Melanosis/genetics , Neurocutaneous Syndromes/drug therapy , Neurocutaneous Syndromes/genetics , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/therapeutic use , Child, Preschool , GTP Phosphohydrolases/genetics , Heterografts/drug effects , Heterografts/pathology , Humans , Male , Melanosis/pathology , Membrane Proteins/genetics , Mice , Mice, Nude , Mutation , Neurocutaneous Syndromes/pathology , Signal Transduction/drug effects , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/therapeutic use , Transcriptome , Tumor Cells, Cultured
17.
Cancer Res ; 74(24): 7260-73, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25336188

ABSTRACT

Oncolytic virus therapy is being evaluated in clinical trials for human glioma. While it is widely assumed that the immune response of the patient to the virus infection limits the utility of the therapy, investigations into the specific cell type(s) involved in this response have been performed using nonspecific pharmacologic inhibitors or allogeneic models with compromised immunity. To identify the immune cells that participate in clearing an oncolytic infection in glioma, we used flow cytometry and immunohistochemistry to immunophenotype an orthotopic glioma model in immunocompetent mice after Myxoma virus (MYXV) administration. These studies revealed a large resident microglia and macrophage population in untreated tumors, and robust monocyte, T-, and NK cell infiltration 3 days after MYXV infection. To determine the role on the clinical utility of MYXV therapy for glioma, we used a combination of knockout mouse strains and specific immunocyte ablation techniques. Collectively, our experiments identify an important role for tumor-resident myeloid cells and overlapping roles for recruited NK and T cells in the clearance and efficacy of oncolytic MYXV from gliomas. Using a cyclophosphamide regimen to achieve lymphoablation prior and during MYXV treatment, we prevented treatment-induced peripheral immunocyte recruitment and, surprisingly, largely ablated the tumor-resident macrophage population. Virotherapy of cyclophosphamide-treated animals resulted in sustained viral infection within the glioma as well as a substantial survival advantage. This study demonstrates that resistance to MYXV virotherapy in syngeneic glioma models involves a multifaceted cellular immune response that can be overcome with cyclophosphamide-mediated lymphoablation.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Myxoma virus/immunology , Oncolytic Virotherapy , Animals , Brain Neoplasms/immunology , Brain Neoplasms/virology , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Flow Cytometry , Glioma/immunology , Glioma/virology , Humans , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Mice , Oncolytic Viruses/immunology , Sirolimus/administration & dosage , Xenograft Model Antitumor Assays
18.
Clin Cancer Res ; 20(22): 5756-67, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25316808

ABSTRACT

PURPOSE: The EGFR and PI3K/mTORC1/2 pathways are frequently altered in glioblastoma (GBM), but pharmacologic targeting of EGFR and PI3K signaling has failed to demonstrate efficacy in clinical trials. Lack of relevant models has rendered it difficult to assess whether targeting these pathways might be effective in molecularly defined subgroups of GBMs. Here, human brain tumor-initiating cell (BTIC) lines with different combinations of endogenous EGFR wild-type, EGFRvIII, and PTEN mutations were used to investigate response to the EGFR inhibitor gefitinib, mTORC1 inhibitor rapamycin, and dual mTORC1/2 inhibitor AZD8055 alone and in combination with temozolomide (TMZ) EXPERIMENTAL DESIGN: In vitro growth inhibition and cell death induced by gefitinib, rapamycin, AZD8055, and TMZ or combinations in human BTICs were assessed by alamarBlue, neurosphere, and Western blotting assays. The in vivo efficacy of AZD8055 was assessed in subcutaneous and intracranial BTIC xenografts. Kaplan-Meier survival studies were performed with AZD8055 and in combination with TMZ. RESULTS: We confirm that gefitinib and rapamycin have modest effects in most BTIC lines, but AZD8055 was highly effective at inhibiting Akt/mTORC2 activity and dramatically reduced the viability of BTICs regardless of their EGFR and PTEN mutational status. Systemic administration of AZD8055 effectively inhibited tumor growth in subcutaneous BTIC xenografts and mTORC1/2 signaling in orthotopic BTIC xenografts. AZD8055 was synergistic with the alkylating agent TMZ and significantly prolonged animal survival. CONCLUSION: These data suggest that dual inhibition of mTORC1/2 may be of benefit in GBM, including the subset of TMZ-resistant GBMs.


Subject(s)
Brain Neoplasms/metabolism , Dacarbazine/analogs & derivatives , Glioblastoma/metabolism , Multiprotein Complexes/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , DNA Methylation , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Disease Models, Animal , Drug Synergism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mutation , PTEN Phosphohydrolase/genetics , Promoter Regions, Genetic , Protein Kinase Inhibitors/administration & dosage , Signal Transduction/drug effects , Temozolomide , Tumor Burden/drug effects , Tumor Burden/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
19.
Clin Cancer Res ; 20(18): 4894-903, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25078279

ABSTRACT

PURPOSE: The current standard of care for glioblastoma (GBM) involves a combination of surgery, radiotherapy, and temozolomide chemotherapy, but this regimen fails to achieve long-term tumor control. Resistance to temozolomide is largely mediated by expression of the DNA repair enzyme MGMT; however, emerging evidence suggests that inactivation of MSH6 and other mismatch repair proteins plays an important role in temozolomide resistance. Here, we investigate endogenous MSH6 mutations in GBM, anaplastic oligodendroglial tumor tissue, and corresponding brain tumor-initiating cell lines (BTIC). EXPERIMENTAL DESIGN: MSH6 sequence and MGMT promoter methylation were determined in human tumor samples and BTICs. Sensitivity to temozolomide was evaluated in vitro using BTICs in the absence and presence of O(6)-benzylguanine to deplete MGMT. The influence of MGMT and MSH6 status on in vivo sensitivity to temozolomide was evaluated using intracranial BTIC xenografts. RESULTS: We identified 11 previously unreported mutations in MSH6 in nine different glioma samples and six paired BTIC lines from adult patients. In addition, MSH6 mutations were documented in three oligodendrogliomas and two treatment-naïve gliomas, both previously unreported findings. These mutations were found to influence the sensitivity of BTICs to temozolomide both in vitro and in vivo, independent of MGMT promoter methylation status. CONCLUSIONS: These data demonstrate that endogenous MSH6 mutations may be present before alkylator therapy and occur in at least two histologic subtypes of adult glial neoplasms, with this report serving as the first to note these mutations in oligodendroglioma. These findings broaden our understanding of the clinical response to temozolomide in gliomas.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Glioblastoma/genetics , Oligodendroglioma/genetics , Promoter Regions, Genetic , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Blotting, Western , Brain Neoplasms/drug therapy , DNA Methylation/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Female , Genotype , Glioblastoma/drug therapy , Humans , Male , Mice , Middle Aged , Mutation , Oligodendroglioma/drug therapy , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Temozolomide , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays , Young Adult
20.
Mol Cancer ; 13: 82, 2014 Apr 13.
Article in English | MEDLINE | ID: mdl-24725816

ABSTRACT

BACKGROUND: Gallbladder carcinoma (GBC) is highly lethal, and effective treatment will require synergistic anti-tumor management. The study is aimed at investigating the oncolytic value of myxoma virus (MYXV) infection against GBC and optimizing MYXV oncolytic efficiency. METHODS: We examined the permissiveness of GBC cell lines to MYXV infection and compared the effects of MYXV on cell viability among GBC and control permissive glioma cells in vitro and in vivo after MYXV + rapamycin (Rap) treatment, which is known to enhance cell permissiveness to MYXV by upregulating p-Akt levels. We also assessed MYXV + hyaluronan (HA) therapy efficiency by examinating Akt activation status, MMP-9 expression, cell viability, and collagen distribution. We further compared hydraulic conductivity, tumor area, and survival of tumor-bearing mice between the MYXV + Rap and MYXV + HA therapeutic regimens. RESULTS: MYXV + Rap treatment could considerably increase the oncolytic ability of MYXV against GBC cell lines in vitro but not against GBC xenografts in vivo. We found higher levels of collagen IV in GBC tumors than in glioma tumors. Diffusion analysis demonstrated that collagen IV could physically hinder MYXV intratumoral distribution. HA-CD44 interplay was found to activate the Akt signaling pathway, which increases oncolytic rates. HA was also found to enhance the MMP-9 secretion, which contributes to collagen IV degradation. CONCLUSIONS: Unlike MYXV + Rap, MYXV + HA therapy significantly enhanced the anti-tumor effects of MYXV in vivo and prolonged survival of GBC tumor-bearing mice. HA may optimize the oncolytic effects of MYXV on GBC via the HA-CD44 interaction which can promote viral infection and diffusion.


Subject(s)
Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/therapy , Oncolytic Virotherapy , Animals , Cell Line, Tumor , Gallbladder Neoplasms/pathology , Gallbladder Neoplasms/virology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronic Acid/administration & dosage , In Vitro Techniques , Mice , Myxoma virus/genetics , Sirolimus/administration & dosage , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL