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










Publication year range
1.
BMC Cancer ; 21(1): 1061, 2021 Sep 26.
Article in English | MEDLINE | ID: mdl-34565342

ABSTRACT

BACKGROUND: Neuroblastoma (NB) patients with MYCN amplification or overexpression respond poorly to current therapies and exhibit extremely poor clinical outcomes. PI3K-mTOR signaling-driven deregulation of protein synthesis is very common in NB and various other cancers that promote MYCN stabilization. In addition, both the MYCN and mTOR signaling axes can directly regulate a common translation pathway that leads to increased protein synthesis and cell proliferation. However, a strategy of concurrently targeting MYCN and mTOR signaling in NB remains unexplored. This study aimed to investigate the therapeutic potential of targeting dysregulated protein synthesis pathways by inhibiting the MYCN and mTOR pathways together in NB. METHODS: Using small molecule/pharmacologic approaches, we evaluated the effects of combined inhibition of MYCN transcription and mTOR signaling on NB cell growth/survival and associated molecular mechanism(s) in NB cell lines. We used two well-established BET (bromodomain extra-terminal) protein inhibitors (JQ1, OTX-015), and a clinically relevant mTOR inhibitor, temsirolimus, to target MYCN transcription and mTOR signaling, respectively. The single agent and combined efficacies of these inhibitors on NB cell growth, apoptosis, cell cycle and neurospheres were assessed using MTT, Annexin-V, propidium-iodide staining and sphere assays, respectively. Effects of inhibitors on global protein synthesis were quantified using a fluorescence-based (FamAzide)-based protein synthesis assay. Further, we investigated the specificities of these inhibitors in targeting the associated pathways/molecules using western blot analyses. RESULTS: Co-treatment of JQ1 or OTX-015 with temsirolimus synergistically suppressed NB cell growth/survival by inducing G1 cell cycle arrest and apoptosis with greatest efficacy in MYCN-amplified NB cells. Mechanistically, the co-treatment of JQ1 or OTX-015 with temsirolimus significantly downregulated the expression levels of phosphorylated 4EBP1/p70-S6K/eIF4E (mTOR components) and BRD4 (BET protein)/MYCN proteins. Further, this combination significantly inhibited global protein synthesis, compared to single agents. Our findings also demonstrated that both JQ1 and temsirolimus chemosensitized NB cells when tested in combination with cisplatin chemotherapy. CONCLUSIONS: Together, our findings demonstrate synergistic efficacy of JQ1 or OTX-015 and temsirolimus against MYCN-driven NB, by dual-inhibition of MYCN (targeting transcription) and mTOR (targeting translation). Additional preclinical evaluation is warranted to determine the clinical utility of targeted therapy for high-risk NB patients.


Subject(s)
Acetanilides/pharmacology , Azepines/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Neuroblastoma/drug therapy , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triazoles/pharmacology , Adaptor Proteins, Signal Transducing/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Down-Regulation , Drug Synergism , Eukaryotic Initiation Factor-4E/drug effects , Eukaryotic Initiation Factor-4E/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Sirolimus/pharmacology , Spheroids, Cellular/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism
2.
Neoplasia ; 23(6): 624-633, 2021 06.
Article in English | MEDLINE | ID: mdl-34107377

ABSTRACT

Amplification of MYCN is a poor prognostic feature in neuroblastoma (NBL) indicating aggressive disease. We and others have shown BET bromodomain inhibitors (BETi) target MYCN indirectly by downregulating its transcription. Here we sought to identify agents that synergize with BETi and to identify biomarkers of resistance. We previously performed a viability screen of ∼1,900 oncology-focused compounds combined with BET bromodomain inhibitors against MYCN-amplified NBL cell lines. Reanalysis of our screening results prominently identified inhibitors of aurora kinase A (AURKAi) to be highly synergistic with BETi. We confirmed the anti-proliferative effects of several BETi+AURKAi combinations in MYCN-amplified NBL cell lines. Compared to single agents, these combinations cooperated to decrease levels of N-myc. We treated both TP53-wild type and mutant, MYCN-amplified cell lines with the BETi JQ1 and the AURKAi Alisertib. The combination had improved efficacy in the TP53-WT context, notably driving apoptosis in both genetic backgrounds. JQ1+Alisertib combination treatment of a MYCN-amplified, TP53-null or TP53-restored genetically engineered mouse model of NBL prolonged survival better than either single agent. This was most profound with TP53 restored, with marked tumor shrinkage and apoptosis induction in response to combination JQ1+Alisertib. BETi+AURKAi in MYCN-amplified NBL, particularly in the context of functional TP53, provided anti-tumor benefits in preclinical models. This combination should be studied more closely in a pediatric clinical trial.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Gene Amplification , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/metabolism , Protein Kinase Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Gene Editing , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Mice , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
3.
Int J Mol Sci ; 22(4)2021 Feb 13.
Article in English | MEDLINE | ID: mdl-33668642

ABSTRACT

Childhood ependymomas are heterogenous chemoresistant neoplasms arising from aberrant stem-like cells. Epigenome deregulation plays a pivotal role in ependymoma pathogenesis, suggesting that epigenetic modifiers hold therapeutic promise against this disease. Bromodomain and extraterminal domain (BET) proteins are epigenome readers of acetylated signals in histones and coactivators for oncogenic and stemness-related transcriptional networks, including MYC/MYCN (Proto-Oncogene, BHLH Transcritpion Factor)-regulated genes. We explored BET inhibition as an anticancer strategy in a panel of pediatric patient-derived ependymoma stem cell models by OTX015-mediated suppression of BET/acetylated histone binding. We found that ependymoma tissues and lines express BET proteins and their targets MYC and MYCN. In vitro, OTX015 reduced cell proliferation by inducing G0/G1-phase accumulation and apoptosis at clinically tolerable doses. Mechanistically, inhibitory p21 and p27 increased in a p53-independent manner, whereas the proliferative driver, phospho-signal transducer and activator of transcription 3 (STAT3), decreased. Upregulation of apoptosis-related proteins and survivin downregulation were correlated with cell line drug sensitivity. Minor alterations of MYC/MYCN expression were reported. In vivo, OTX015 significantly improved survival in 2/3 orthotopic ependymoma models. BET proteins represent promising targets for pharmaceutical intervention with OTX015 against ependymoma. The identification of predictive determinants of sensitivity may help identify ependymoma molecular subsets more likely to benefit from BET inhibitor therapies.


Subject(s)
Acetanilides/pharmacology , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Ependymoma/drug therapy , Heterocyclic Compounds, 3-Ring/pharmacology , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Cell Line, Tumor , Ependymoma/metabolism , Ependymoma/pathology , Humans , Male , Mice , Mice, Nude , N-Myc Proto-Oncogene Protein/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/metabolism , Xenograft Model Antitumor Assays
4.
Cancer Res ; 81(7): 1627-1632, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33509943

ABSTRACT

Effective treatment of pediatric solid tumors has been hampered by the predominance of currently "undruggable" driver transcription factors. Improving outcomes while decreasing the toxicity of treatment necessitates the development of novel agents that can directly inhibit or degrade these elusive targets. MYCN in pediatric neural-derived tumors, including neuroblastoma and medulloblastoma, is a paradigmatic example of this problem. Attempts to directly and specifically target MYCN have failed due to its similarity to MYC, the unstructured nature of MYC family proteins in their monomeric form, the lack of an understanding of MYCN-interacting proteins and ability to test their relevance in vivo, the inability to obtain structural information on MYCN protein complexes, and the challenges of using traditional small molecules to inhibit protein-protein or protein-DNA interactions. However, there is now promise for directly targeting MYCN based on scientific and technological advances on all of these fronts. Here, we discuss prior challenges and the reasons for renewed optimism in directly targeting this "undruggable" transcription factor, which we hope will lead to improved outcomes for patients with pediatric cancer and create a framework for targeting driver oncoproteins regulating gene transcription.


Subject(s)
Antineoplastic Agents/isolation & purification , Drug Resistance, Neoplasm , N-Myc Proto-Oncogene Protein/physiology , Neoplasms/drug therapy , Therapies, Investigational , Age of Onset , Antineoplastic Agents/history , Antineoplastic Agents/therapeutic use , Child , Drug Discovery/history , Drug Discovery/methods , Drug Discovery/trends , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor/history , Drug Screening Assays, Antitumor/methods , Drug Screening Assays, Antitumor/trends , Gene Expression Regulation, Neoplastic/drug effects , History, 20th Century , History, 21st Century , Humans , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/genetics , Neoplasms/epidemiology , Neoplasms/genetics , Therapies, Investigational/history , Therapies, Investigational/methods , Therapies, Investigational/trends
5.
Int J Mol Sci ; 21(21)2020 Nov 05.
Article in English | MEDLINE | ID: mdl-33167327

ABSTRACT

Resistance to androgen-receptor (AR) directed therapies is, among other factors, associated with Myc transcription factors that are involved in development and progression of many cancers. Overexpression of N-Myc protein in prostate cancer (PCa) leads to its transformation to advanced neuroendocrine prostate cancer (NEPC) that currently has no approved treatments. N-Myc has a short half-life but acts as an NEPC stimulator when it is stabilized by forming a protective complex with Aurora A kinase (AURKA). Therefore, dual-inhibition of N-Myc and AURKA would be an attractive therapeutic avenue for NEPC. Following our computer-aided drug discovery approach, compounds exhibiting potent N-Myc specific inhibition and strong anti-proliferative activity against several N-Myc driven cell lines, were identified. Thereafter, we have developed dual inhibitors of N-Myc and AURKA through structure-based drug design approach by merging our novel N-Myc specific chemical scaffolds with fragments of known AURKA inhibitors. Favorable binding modes of the designed compounds to both N-Myc and AURKA target sites have been predicted by docking. A promising lead compound, 70812, demonstrated low-micromolar potency against both N-Myc and AURKA in vitro assays and effectively suppressed NEPC cell growth.


Subject(s)
Antineoplastic Agents/isolation & purification , Aurora Kinase A/antagonists & inhibitors , Carcinoma, Neuroendocrine/drug therapy , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cells, Cultured , Drug Discovery/methods , Drug Screening Assays, Antitumor , Drugs, Investigational/chemistry , Drugs, Investigational/isolation & purification , Drugs, Investigational/pharmacology , Humans , Male , Models, Molecular , Molecular Docking Simulation , Molecular Targeted Therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/isolation & purification , Protein Kinase Inhibitors/pharmacology , Receptors, Androgen/metabolism
6.
Cancer Res ; 80(16): 3424-3435, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32595135

ABSTRACT

Noninvasive early indicators of treatment response are crucial to the successful delivery of precision medicine in children with cancer. Neuroblastoma is a common solid tumor of young children that arises from anomalies in neural crest development. Therapeutic approaches aiming to destabilize MYCN protein, such as small-molecule inhibitors of Aurora A and mTOR, are currently being evaluated in early phase clinical trials in children with high-risk MYCN-driven disease, with limited ability to evaluate conventional pharmacodynamic biomarkers of response. T1 mapping is an MRI scan that measures the proton spin-lattice relaxation time T1. Using a multiparametric MRI-pathologic cross-correlative approach and computational pathology methodologies including a machine learning-based algorithm for the automatic detection and classification of neuroblasts, we show here that T1 mapping is sensitive to the rich histopathologic heterogeneity of neuroblastoma in the Th-MYCN transgenic model. Regions with high native T1 corresponded to regions dense in proliferative undifferentiated neuroblasts, whereas regions characterized by low T1 were rich in apoptotic or differentiating neuroblasts. Reductions in tumor-native T1 represented a sensitive biomarker of response to treatment-induced apoptosis with two MYCN-targeted small-molecule inhibitors, Aurora A kinase inhibitor alisertib (MLN8237) and mTOR inhibitor vistusertib (AZD2014). Overall, we demonstrate the potential of T1 mapping, a scan readily available on most clinical MRI scanners, to assess response to therapy and guide clinical trials for children with neuroblastoma. The study reinforces the potential role of MRI-based functional imaging in delivering precision medicine to children with neuroblastoma. SIGNIFICANCE: This study shows that MRI-based functional imaging can detect apoptotic responses to MYCN-targeted small-molecule inhibitors in a genetically engineered murine model of MYCN-driven neuroblastoma.


Subject(s)
Benzamides/therapeutic use , Morpholines/therapeutic use , Multiparametric Magnetic Resonance Imaging/methods , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Neuroblastoma/diagnostic imaging , Neuroblastoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Algorithms , Animals , Azepines/therapeutic use , Child , Female , Humans , Machine Learning , Male , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/pathology , Precision Medicine/methods , TOR Serine-Threonine Kinases/antagonists & inhibitors , Time Factors , Treatment Outcome
7.
Anticancer Agents Med Chem ; 20(13): 1613-1625, 2020.
Article in English | MEDLINE | ID: mdl-32329693

ABSTRACT

BACKGROUND: MYCN amplification is a prognostic biomarker associated with poor prognosis of neuroblastoma in children. The overall survival of children with MYCN-amplified neuroblastoma has only marginally improved within the last 20 years. The Bromodomain and Extra-Terminal motif (BET) inhibitor, JQ1, has been shown to downregulate MYCN in neuroblastoma cells. OBJECTIVE: To determine if JQ1 downregulation of MYCN in neuroblastomas can offer a target- specific therapy for this, difficult to treat, pediatric cancer. METHODS: Since MYCN-amplified neuroblastoma accounts for as much as 40 to 50 percent of all high-risk cases, we compared the effect of JQ1 on both MYCN-amplified and non-MYCN-amplified neuroblastoma cell lines and investigated its mechanism of action. RESULTS: In this study, we show that JQ1 can specifically target MYCN for downregulation, though this effect is not specific to only MYCN-amplified cells. And although we can confirm that the loss of MYCN alone can induce apoptosis, the exogenous rescue of MYCN expression can abrogate much of this cytotoxicity. More fascinating, however, was the discovery that the JQ1-induced knockdown of MYCN, which led to the loss of the human double minute 2 homolog (HDM2) protein, also led to the accumulation of tumor protein 53 (also known as TP53 or p53), which ultimately induced apoptosis. Likewise, the knockdown of p53 also blunted the cytotoxic effects of JQ1. CONCLUSION: These data suggest a mechanism of action for JQ1 cytotoxicity in neuroblastomas and offer a possible prognostic target for determining its efficacy as a therapeutic.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Triazoles/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Azepines/chemical synthesis , Azepines/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Humans , Molecular Structure , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/chemistry , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
8.
Nucleic Acid Ther ; 30(4): 237-248, 2020 08.
Article in English | MEDLINE | ID: mdl-32240058

ABSTRACT

Neuroblastoma (NB) is the most common solid tumor in childhood. Twenty percent of patients display MYCN amplification, which indicates a very poor prognosis. MYCN is a highly specific target for an NB tumor therapy as MYCN expression is absent or very low in most normal cells, while, as a transcription factor, it regulates many essential cell activities in tumor cells. We aim to develop a therapy for NB based on MYCN silencing by short interfering RNA (siRNA) molecules, which can silence target genes by RNA interference (RNAi), a naturally occurring method of gene silencing. It has been shown previously that MYCN silencing can induce apoptosis and differentiation in MYCN amplified NB. In this article, we have demonstrated that siRNA-mediated silencing of MYCN in MYCN-amplified NB cells induced neurogenesis in NB cells, whereas retinoic acid (RA) treatment did not. RA can differentiate NB cells and is used for treatment of residual disease after surgery or chemotherapy, but resistance can develop. In addition, MYCN siRNA treatment suppressed growth in a MYCN-amplified NB cell line more than that by RA. Our result suggests that gene therapy using RNAi targeting MYCN can be a novel therapy toward MYCN-amplified NB that have complete or partial resistance toward RA.


Subject(s)
Gene Silencing/drug effects , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/drug therapy , RNA, Small Interfering/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Neuroblastoma/genetics , Neuroblastoma/pathology , Neurogenesis/drug effects , RNA Interference/drug effects , RNA, Small Interfering/genetics , Tretinoin/adverse effects , Tretinoin/pharmacology
9.
Cancer Res ; 79(24): 6166-6177, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31615807

ABSTRACT

Approximately half of high-risk neuroblastoma is characterized by MYCN amplification. N-Myc promotes tumor progression by inducing cell growth and inhibiting differentiation. MYCN has also been shown to play an active role in mitochondrial metabolism, but this relationship is not well understood. Although N-Myc is a known driver of the disease, it remains a target for which no therapeutic drug exists. Here, we evaluated a novel MYCN-specific antigene PNA oligonucleotide (BGA002) in MYCN-amplified (MNA) or MYCN-expressing neuroblastoma and investigated the mechanism of its antitumor activity. MYCN mRNA and cell viability were reduced in a broad set of neuroblastoma cell lines following BGA002 treatment. Furthermore, BGA002 decreased N-Myc protein levels and apoptosis in MNA neuroblastoma. Analysis of gene expression data from patients with neuroblastoma revealed that MYCN was associated with increased reactive oxygen species (ROS), downregulated mitophagy, and poor prognosis. Inhibition of MYCN caused profound mitochondrial damage in MNA neuroblastoma cells through downregulation of the mitochondrial molecular chaperone TRAP1, which subsequently increased ROS. Correspondingly, inhibition of MYCN reactivated mitophagy. Systemic administration of BGA002 downregulated N-Myc and TRAP1, with a concomitant decrease in MNA neuroblastoma xenograft tumor weight. In conclusion, this study highlights the role of N-Myc in blocking mitophagy in neuroblastoma and in conferring protection to ROS in mitochondria through upregulation of TRAP1. BGA002 is a potently improved MYCN-specific antigene oligonucleotide that reverts N-Myc-dysregulated mitochondrial pathways, leading to loss of the protective effect of N-Myc against mitochondrial ROS. SIGNIFICANCE: A second generation antigene peptide oligonucleotide targeting MYCN induces mitochondrial damage and inhibits growth of MYCN-amplified neuroblastoma cells.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Mitochondria/drug effects , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/pathology , Peptide Nucleic Acids/pharmacology , Adolescent , Adult , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Child, Preschool , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Infant , Infant, Newborn , Kaplan-Meier Estimate , Male , Mice , Mitochondria/metabolism , Mitochondria/pathology , Mitophagy/drug effects , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/mortality , Peptide Nucleic Acids/genetics , Peptide Nucleic Acids/therapeutic use , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays , Young Adult
10.
Bioorg Chem ; 91: 103165, 2019 10.
Article in English | MEDLINE | ID: mdl-31419642

ABSTRACT

MicroRNAs are a ubiquitous class of non-coding RNAs able to regulate gene expression in diverse biological processes. Widespread miRNAs deregulation was reported in numerous diseases including cancer, with several miRNAs playing oncogenic and/or tumor suppressive role by targeting multiple mRNAs simultaneously. Based on these findings, miRNAs have emerged as promising therapeutic tools for cancer treatment. Herein, for the first time, peptide nucleic acids (PNAs) were studied to develop a new class of molecules able to target 3'UTR on MYCN mRNA without a fully complementary base pairing sequence (as miRNAs). For our proof of concept study we have selected as a model the miRNA-34a, which acts as a tumor suppressor in a number of cancers including neuroblastoma. In particular, miRNA-34a is a direct regulator of MYCN oncogene, whose overexpression is a prominent biomarker for the highly aggressive neuroblastoma phenotype. The design and synthesis of three PNA-based oligomers of different length was described, and their interaction with two binding sites on the target MYCN mRNA was investigated by molecular dynamics simulation, and spectroscopic techniques (CD, UV). Intake assay and confocal microscopy of PNA sequences were also carried out in vitro on neuroblastoma Kelly cells. Despite the presence of multiple mismatches, the PNA/RNA hetero duplexes retain very interesting features in terms of stability, affinity as well as of cellular uptake.


Subject(s)
Cell Proliferation , MicroRNAs/genetics , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Neuroblastoma/drug therapy , Peptide Nucleic Acids/pharmacology , RNA, Messenger/antagonists & inhibitors , Computational Chemistry , Humans , Microscopy, Confocal , Molecular Dynamics Simulation , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Peptide Nucleic Acids/chemical synthesis , RNA, Messenger/genetics , Tumor Cells, Cultured
11.
Cell Physiol Biochem ; 53(1): 258-280, 2019.
Article in English | MEDLINE | ID: mdl-31313541

ABSTRACT

BACKGROUND/AIMS: Although neuroblastoma is a heterogeneous cancer, a substantial portion overexpresses CD71 (transferrin receptor 1) and MYCN. This study provides a mechanistically driven rationale for a combination therapy targeting neuroblastomas that doubly overexpress or have amplified CD71 and MYCN. For this subset, CD71 was targeted by its natural ligand, gambogic acid (GA), and MYCN was targeted with an HDAC inhibitor, vorinostat. A combination of GA and vorinostat was then tested for efficacy in cancer and non-cancer cells. METHODS: Microarray analysis of cohorts of neuroblastoma patients indicated a subset of neuroblastomas overexpressing both CD71 and MYCN. The viability with proliferation changes were measured by MTT and colony formation assays in neuroblastoma cells. Transfection with CD71 or MYCN along with quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were used to detect expression changes. For pathway analysis, gene ontology (GO) and Protein-protein interaction analyses were performed to evaluate the potential mechanisms of GA and vorinostat in treated cells. RESULTS: For both GA and vorinostat, their pathways were explored for specificity and dependence on their targets for efficacy. For GA-treated cells, the viability/proliferation loss due to GA was dependent on the expression of CD71 and involved activation of caspase-3 and degradation of EGFR. It relied on the JNK-IRE1-mTORC1 pathway. The drug vorinostat also reduced cell viability/proliferation in the treated cells and this was dependent on the presence of MYCN as MYCN siRNA transfection led to a blunting of vorinostat efficacy and conversely, MYCN overexpression improved the vorinostat potency in those cells. Vorinostat inhibition of MYCN led to an increase of the pro-apoptotic miR183 levels and this, in turn, reduced the viability/proliferation of these cells. The combination treatment with GA and vorinostat synergistically reduced cell survival in the MYCN and CD71 overexpressing tumor cells. The same treatment had no effect or minimal effect on HEK293 and HEF cells used as models of non-cancer cells. CONCLUSION: A combination therapy with GA and vorinostat may be suitable for MYCN and CD71 overexpressing neuroblastomas.


Subject(s)
Antigens, CD , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Delivery Systems , N-Myc Proto-Oncogene Protein , Neuroblastoma , Receptors, Transferrin , Antigens, CD/genetics , Antigens, CD/metabolism , Caspase 3/genetics , Caspase 3/metabolism , HEK293 Cells , Humans , MicroRNAs/biosynthesis , MicroRNAs/genetics , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptors, Transferrin/antagonists & inhibitors , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Vorinostat/pharmacology , Xanthones/pharmacology
12.
Cell Prolif ; 52(3): e12577, 2019 May.
Article in English | MEDLINE | ID: mdl-30724400

ABSTRACT

OBJECTIVES: This study aims to explore the roles of N-myc and caspase-8 in TRAIL-resistant IMR-32 cells which exhibit MYCN oncogene amplification and lack caspase-8 expression. MATERIALS AND METHODS: We established N-myc-downregulated IMR-32 cells using shRNA lentiviral particles targeting N-myc and examined the effect the N-myc inhibition on TRAIL susceptibility in human neuroblastoma IMR-32 cells expressing caspase-8. RESULTS: Cisplatin treatment in IMR-32 cells increased the expression of death receptor 5 (DR5; TRAIL-R2), but not other receptors, via downregulation of NF-κB activity. However, the cisplatin-mediated increase in DR5 failed to induce cell death following TRAIL treatment. Furthermore, interferon (IFN)-γ pretreatment increased caspase-8 expression in IMR-32 cells, but cisplatin failed to trigger TRAIL cytotoxicity. We downregulated N-myc expression in IMR-32 cells using N-myc-targeting shRNA. These cells showed decreased growth rate and Bcl-2 expression accompanied by a mild collapse in the mitochondrial membrane potential as compared with those treated with scrambled shRNA. TRAIL treatment in N-myc-negative cells expressing caspase-8 following IFN-γ treatment significantly triggered apoptotic cell death. Concurrent treatment with cisplatin enhanced TRAIL-mediated cytotoxicity, which was abrogated by an additional pretreatment with DR5:Fc chimera protein. CONCLUSIONS: N-myc and caspase-8 expressions are involved in TRAIL susceptibility in IMR-32 cells, and the combination of treatment with cisplatin and TRAIL may serve as a promising strategy for the development of therapeutics against neuroblastoma that is controlled by N-myc and caspase-8 expression.


Subject(s)
Caspase 8/genetics , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/therapy , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cisplatin/administration & dosage , Gene Expression , Genes, myc , Humans , Interferon-gamma/administration & dosage , MAP Kinase Signaling System , NF-kappa B/genetics , NF-kappa B/metabolism , Neuroblastoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Recombinant Proteins/administration & dosage
13.
J Biochem Mol Toxicol ; 33(5): e22292, 2019 May.
Article in English | MEDLINE | ID: mdl-30672053

ABSTRACT

The pathogenetic mechanisms of retinoblastoma are still not yet fully elucidated, putting limits to efficacious treatment. Crocin is the main component of saffron, which exhibits significant antitumorigenic properties. The aim of this paper is to investigate the effect of crocin on retinoblastoma. The effects of crocin on the proliferation of human retinoblastoma cells were determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, cell number assay, and colony formation assay. Cell apoptosis induced by crocin was measured by flow cytometry analysis. Cleaved poly(ADP-ribose) polymerase and cleaved caspase-3 were tested by western blot analysis. The expression levels of MYCN were assessed by western blot and quantitative polymerase chain reaction and the stability of MYCN messenger RNA was determined by in vitro RNA degradation assays. We found that crocin significantly inhibited the cell proliferation and clonogenicity and induced cell apoptosis in Y79 and WERI-RB-1 cells. In addition, crocin treatment significantly reduced the expression and the stability of MYCN. Besides, overexpression of MYCN rescued the inhibitory effect of crocin in Y79 cells. Our findings suggest that crocin exhibits antitumorigenic effects in human retinoblastoma cell lines through a MYCN-dependent manner, which may provide guidance to logical therapeutic designs in prevention and treatment of retinoblastoma.


Subject(s)
Apoptosis/drug effects , Carotenoids/pharmacology , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , N-Myc Proto-Oncogene Protein/biosynthesis , Retinoblastoma/drug therapy , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Protein Stability/drug effects , Retinoblastoma/metabolism , Retinoblastoma/pathology
14.
Cell Death Dis ; 9(2): 220, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445162

ABSTRACT

Heightened aerobic glycolysis and glutaminolysis are characteristic metabolic phenotypes in cancer cells. Neuroblastoma (NBL), a devastating pediatric cancer, is featured by frequent genomic amplification of MYCN, a member of the Myc oncogene family that is primarily expressed in the early stage of embryonic development and required for neural crest development. Here we report that an enriched glutaminolysis gene signature is associated with MYCN amplification in children with NBL. The partial knockdown of MYCN suppresses glutaminolysis in NBL cells. Conversely, forced overexpression of MYCN in neural crest progenitor cells enhances glutaminolysis. Importantly, glutaminolysis induces oxidative stress by producing reactive oxygen species (ROS), rendering NBL cells sensitive to ROS augmentation. Through a small-scale metabolic-modulator screening, we have found that dimethyl fumarate (DMF), a Food and Drug Administration-approved drug for multiple sclerosis, suppresses NBL cell proliferation in vitro and tumor growth in vivo. DMF suppresses NBL cell proliferation through inducing ROS and subsequently suppressing MYCN expression, which is rescued by an ROS scavenger. Our findings suggest that the metabolic modulation and ROS augmentation could be used as novel strategies in treating NBL and other MYC-driven cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Dimethyl Fumarate/pharmacology , Gene Expression Regulation, Neoplastic , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Allografts , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Dimethyl Fumarate/antagonists & inhibitors , Drug Screening Assays, Antitumor , Free Radical Scavengers/pharmacology , Glutamine/agonists , Glutamine/antagonists & inhibitors , Glutamine/metabolism , Glycolysis/drug effects , Glycolysis/genetics , Humans , Mice , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/metabolism , Neural Crest/drug effects , Neural Crest/metabolism , Neural Crest/pathology , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Signal Transduction
15.
Sci Rep ; 7(1): 13612, 2017 10 19.
Article in English | MEDLINE | ID: mdl-29051534

ABSTRACT

Neuroblastoma is the most common extra-cranial solid tumor in children. Its broad spectrum of clinical outcomes reflects the underlying inherent cellular heterogeneity. As current treatments often do not lead to tumor eradication, there is a need to better define therapy-resistant neuroblastoma and to identify new modulatory molecules. To this end, we performed the first comprehensive flow cytometric characterization of surface molecule expression in neuroblastoma cell lines. Exploiting an established clustering algorithm (SPADE) for unbiased visualization of cellular subsets, we conducted a multiwell screen for small molecule modulators of neuroblastoma phenotype. In addition to SH-SY5Y cells, the SH-EP, BE(2)-M17 and Kelly lines were included in follow-up analysis as in vitro models of neuroblastoma. A combinatorial detection of glycoprotein epitopes (CD15, CD24, CD44, CD57, TrkA) and the chemokine receptor CXCR4 (CD184) enabled the quantitative identification of SPADE-defined clusters differentially responding to small molecules. Exposure to bone morphogenetic protein (BMP)-4 was found to enhance a TrkAhigh/CD15-/CD184- neuroblastoma cellular subset, accompanied by a reduction in doublecortin-positive neuroblasts and of NMYC protein expression in SH-SY5Y cells. Beyond yielding novel marker candidates for studying neuroblastoma pathology, our approach may provide tools for improved pharmacological screens towards developing novel avenues of neuroblastoma diagnosis and treatment.


Subject(s)
Antigens, Surface/metabolism , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Small Molecule Libraries/pharmacology , Cell Line, Tumor , Cluster Analysis , Humans , Lewis X Antigen/metabolism , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA Interference , RNA, Small Interfering/metabolism , Receptor, trkA/metabolism
16.
Mol Cell ; 64(3): 493-506, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27773673

ABSTRACT

MYCN amplification in human cancers predicts poor prognosis and resistance to therapy. However, pharmacological strategies that directly target N-Myc, the protein encoded by MYCN, remain elusive. Here, we identify a molecular mechanism responsible for reciprocal activation between Polo-like kinase-1 (PLK1) and N-Myc. PLK1 specifically binds to the SCFFbw7 ubiquitin ligase, phosphorylates it, and promotes its autopolyubiquitination and proteasomal degradation, counteracting Fbw7-mediated degradation of N-Myc and additional substrates, including cyclin E and Mcl1. Stabilized N-Myc in turn directly activates PLK1 transcription, constituting a positive feedforward regulatory loop that reinforces Myc-regulated oncogenic programs. Inhibitors of PLK1 preferentially induce potent apoptosis of MYCN-amplified tumor cells from neuroblastoma and small cell lung cancer and synergistically potentiate the therapeutic efficacies of Bcl2 antagonists. These findings reveal a PLK1-Fbw7-Myc signaling circuit that underlies tumorigenesis and validate PLK1 inhibitors, alone or with Bcl2 antagonists, as potential effective therapeutics for MYC-overexpressing cancers.


Subject(s)
Brain Neoplasms/genetics , Cell Cycle Proteins/genetics , F-Box Proteins/genetics , Feedback, Physiological , Gene Expression Regulation, Neoplastic , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Drug Synergism , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Humans , Mice, Nude , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/mortality , Neuroblastoma/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pteridines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sulfonamides/pharmacology , Survival Analysis , Transcription, Genetic , Tumor Burden/drug effects , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
17.
Dis Model Mech ; 9(9): 941-52, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27483357

ABSTRACT

The first-in-class inhibitor of ALK, c-MET and ROS1, crizotinib (Xalkori), has shown remarkable clinical efficacy in treatment of ALK-positive non-small cell lung cancer. However, in neuroblastoma, activating mutations in the ALK kinase domain are typically refractory to crizotinib treatment, highlighting the need for more potent inhibitors. The next-generation ALK inhibitor PF-06463922 is predicted to exhibit increased affinity for ALK mutants prevalent in neuroblastoma. We examined PF-06463922 activity in ALK-driven neuroblastoma models in vitro and in vivo In vitro kinase assays and cell-based experiments examining ALK mutations of increasing potency show that PF-06463922 is an effective inhibitor of ALK with greater activity towards ALK neuroblastoma mutants. In contrast to crizotinib, single agent administration of PF-06463922 caused dramatic tumor inhibition in both subcutaneous and orthotopic xenografts as well as a mouse model of high-risk neuroblastoma driven by Th-ALK(F1174L)/MYCN Taken together, our results suggest PF-06463922 is a potent inhibitor of crizotinib-resistant ALK mutations, and highlights an important new treatment option for neuroblastoma patients.


Subject(s)
Lactams, Macrocyclic/therapeutic use , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Neuroblastoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Aminopyridines , Anaplastic Lymphoma Kinase , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Clinical Trials as Topic , Crizotinib , Lactams , Lactams, Macrocyclic/pharmacology , Mice, Inbred BALB C , Mice, Nude , Mutation/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/pathology , PC12 Cells , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Xenograft Model Antitumor Assays
18.
Hum Vaccin Immunother ; 12(9): 2257-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27096896

ABSTRACT

Amplification and concomitant overexpression of the MYCN oncogene is a frequent event in many malignancies including the childhood tumors, neuroblastoma and medulloblastoma. MYCN is only expressed in a defined time frame during early developmental processes, (1) which is beneficial for approaches combatting tumor-specific MYCN. However, MYCN is a transcription factors that was considered a poor drug target, until recent approaches suggested that down-regulation of MYCN could be possible by indirect targeting using Aurora kinase inhibitors or BET inhibitors. These concepts were proven using preclinical models (2-6) and are now entering clinical trials.


Subject(s)
Cancer Vaccines/therapeutic use , Medulloblastoma/therapy , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , Neuroblastoma/therapy , Animals , Aurora Kinases/antagonists & inhibitors , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Nerve Tissue Proteins/antagonists & inhibitors
19.
Sensors (Basel) ; 16(3): 290, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26927112

ABSTRACT

Liposome-based drug delivery systems hold great potential for cancer therapy. The aim of this study was to design a nanodevice for targeted anchoring of liposomes (with and without cholesterol) with encapsulated anticancer drugs and antisense N-myc gene oligonucleotide attached to its surface. To meet this main aim, liposomes with encapsulated doxorubicin, ellipticine and etoposide were prepared. They were further characterized by measuring their fluorescence intensity, whereas the encapsulation efficiency was estimated to be 16%. The hybridization process of individual oligonucleotides forming the nanoconstruct was investigated spectrophotometrically and electrochemically. The concentrations of ellipticine, doxorubicin and etoposide attached to the nanoconstruct in gold nanoparticle-modified liposomes were found to be 14, 5 and 2 µg·mL(-1), respectively. The study succeeded in demonstrating that liposomes are suitable for the transport of anticancer drugs and the antisense oligonucleotide, which can block the expression of the N-myc gene.


Subject(s)
DNA, Antisense/therapeutic use , Drug Delivery Systems , Magnetite Nanoparticles/chemistry , Neoplasms/drug therapy , DNA, Antisense/chemistry , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Ellipticines/chemistry , Ellipticines/therapeutic use , Etoposide/chemistry , Etoposide/therapeutic use , Fluorescence , Gold/chemistry , Humans , Liposomes/chemistry , Liposomes/therapeutic use , Magnetite Nanoparticles/therapeutic use , N-Myc Proto-Oncogene Protein/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...