pyrimidinones has been researched along with Neuroblastoma* in 37 studies
37 other study(ies) available for pyrimidinones and Neuroblastoma
Article | Year |
---|---|
FGFR2 loss sensitizes MYCN-amplified neuroblastoma CHP134 cells to CHK1 inhibitor-induced apoptosis.
Checkpoint kinase 1 (CHK1) plays a key role in genome surveillance and integrity throughout the cell cycle. Selective inhibitors of CHK1 (CHK1i) are undergoing clinical evaluation for various human malignancies, including neuroblastoma. In this study, one CHK1i-sensitive neuroblastoma cell line, CHP134, was investigated, which characteristically carries MYCN amplification and a chromosome deletion within the 10q region. Among several cancer-related genes in the chromosome 10q region, mRNA expression of fibroblast growth factor receptor 2 (FGFR2) was altered in CHP134 cells and associated with an unfavorable prognosis of patients with neuroblastoma. Induced expression of FGFR2 in CHP134 cells reactivated downstream MEK/ERK signaling and resulted in cells resistant to CHK1i-mediated cell growth inhibition. Consistently, the MEK1/2 inhibitor, trametinib, potentiated CHK1 inhibitor-mediated cell death in these cells. These results suggested that FGFR2 loss might be prone to highly effective CHK1i treatment. In conclusion, extreme cellular dependency of ERK activation may imply a possible application for the MEK1/2 inhibitor, either as a single inhibitor or in combination with CHK1i in MYCN-amplified neuroblastomas. Topics: Apoptosis; Cell Line, Tumor; Checkpoint Kinase 1; Drug Resistance, Neoplasm; Drug Synergism; Gene Amplification; Humans; MAP Kinase Kinase 1; MAP Kinase Kinase 2; MAP Kinase Signaling System; N-Myc Proto-Oncogene Protein; Neuroblastoma; Prognosis; Protein Kinase Inhibitors; Pyridones; Pyrimidinones; Receptor, Fibroblast Growth Factor, Type 2; RNA, Messenger | 2022 |
Trametinib for a BRAF G469A missense mutation in a neuroblastoma unveiled by liquid biopsy.
Topics: Antineoplastic Combined Chemotherapy Protocols; Humans; Liquid Biopsy; Mutation; Mutation, Missense; Neuroblastoma; Protein Kinase Inhibitors; Proto-Oncogene Proteins B-raf; Pyridones; Pyrimidinones | 2022 |
Phosphodiesterase 5 inhibitor mirodenafil ameliorates Alzheimer-like pathology and symptoms by multimodal actions.
Alzheimer's disease (AD) pathology is associated with complex interactions among multiple factors, involving an intertwined network of various signaling pathways. The polypharmacological approach is an emerging therapeutic strategy that has been proposed to overcome the multifactorial nature of AD by targeting multiple pathophysiological factors including amyloid-β (Aβ) and phosphorylated tau. We evaluated a blood-brain barrier penetrating phosphodiesterase 5 (PDE5) inhibitor, mirodenafil (5-ethyl-2-7-n-propyl-3,5-dihydrro-4H-pyrrolo[3,2-d]pyrimidin-4-one), for its therapeutic effects on AD with polypharmacological properties.. To evaluate the potential of mirodenafil as a disease-modifying AD agent, mirodenafil was administered to test its effects on the cognitive behaviors of the APP-C105 AD mouse model using the Morris water maze and passive avoidance tests. To investigate the mechanisms of action that underlie the beneficial disease-modifying effects of mirodenafil, human neuroblastoma SH-SY5Y cells and mouse hippocampal HT-22 cells were used to show mirodenafil-induced alterations associated with the cyclic guanosine monophosphate (cGMP)/cGMP-dependent protein kinase (PKG)/cAMP-responsive element-binding protein (CREB) pathway, apoptotic cell death, tau phosphorylation, amyloidogenesis, the autophagy-lysosome pathway, glucocorticoid receptor (GR) transcriptional activity, and the Wnt/β-catenin signaling.. Here, mirodenafil is demonstrated to improve cognitive behavior in the APP-C105 mouse model. Mirodenafil not only reduced the Aβ and phosphorylated tau burdens in vivo, but also ameliorated AD pathology induced by Aβ through the modulation of the cGMP/PKG/CREB signaling pathway, glycogen synthase kinase 3β (GSK-3β) activity, GR transcriptional activity, and the Wnt/β-catenin signaling in neuronal cells. Interestingly, homodimerization and nuclear localization of GR were inhibited by mirodenafil, but not by other PDE5 inhibitors. In addition, only mirodenafil reduced the expression levels of the Wnt antagonist Dickkopf-1 (Dkk-1), thus activating the Wnt/β-catenin signaling.. These findings strongly suggest that the PDE5 inhibitor mirodenafil shows promise as a potential polypharmacological drug candidate for AD treatment, acting on multiple key signaling pathways involved in amyloid deposition, phosphorylated tau burden, the cGMP/PKG/CREB pathway, GSK-3β kinase activity, GR signaling, and the Wnt/β-catenin signaling. Mirodenafil administration to the APP-C105 AD mouse model also improved cognitive behavior, demonstrating the potential of mirodenafil as a polypharmacological AD therapeutic agent. Topics: Alzheimer Disease; Amyloid beta-Peptides; Animals; beta Catenin; Cyclic GMP; Disease Models, Animal; Glycogen Synthase Kinase 3 beta; Humans; Mice; Neuroblastoma; Phosphodiesterase 5 Inhibitors; Phosphorylation; Pyrimidinones; Sulfonamides; tau Proteins | 2022 |
Catastrophic ATP loss underlies a metabolic combination therapy tailored for
Topics: Animals; Antineoplastic Combined Chemotherapy Protocols; Apoptosis; Basigin; Cell Line, Tumor; Cell Proliferation; Electron Transport Complex I; Gene Amplification; Humans; Mice; Mitochondria; Monocarboxylic Acid Transporters; N-Myc Proto-Oncogene Protein; Neuroblastoma; Phenformin; Pyrimidinones; Symporters; Thiophenes; Xenograft Model Antitumor Assays | 2021 |
Anti-relapse effect of trametinib on a local minimal residual disease neuroblastoma mouse model.
We reported the in vitro and in vivo anti-tumor effects of trametinib, an MEK inhibitor, on neuroblastoma. However, long-term trametinib administration for bulky tumors failed to prevent local relapse. In this study, we established a local minimal residual disease (L-MRD) model to develop an optimal clinical protocol.. We prepared an l-MRD model by implanting neuroblastoma cells (SK-N-AS) into the renal capsule of nude mice with total tumorectomy or sham operation 14 days later. These mice received post-operative administration of trametinib or vehicle for eight weeks. Relapse was measured once weekly. Flow cytometry was performed with SK-N-AS cells treated by trametinib.. Tumorectomy+trametinib dramatically suppressed relapse, and all mice survived during trametinib administration, while other treatments failed to suppress relapse. The survival rates for other groups were 20% in sham+trametinib, 17% in tumorectomy+vehicle, and 0% in sham+vehicle. Relapse occurred in the tumorectomy+trametinib group after withdrawal of trametinib administration. Flow cytometry revealed G1 arrest in SK-N-AS cells treated with trametinib.. These findings suggested that trametinib was able to suppress relapse from minimal residual tumor cells. Therefore, we propose that trametinib be administered as an option for maintenance therapy after surgical and chemotherapeutic treatments for neuroblastoma in future clinical protocols. Topics: Animals; Cell Line, Tumor; Mice; Mice, Nude; Neoplasm Recurrence, Local; Neoplasm, Residual; Neuroblastoma; Protein Kinase Inhibitors; Pyridones; Pyrimidinones | 2021 |
The synergistic antitumor effect of combination therapy with a MEK inhibitor and YAP inhibitor on pERK-positive neuroblastoma.
We previously reported the in vitro and in vivo antitumor effects of trametinib, a MEK inhibitor, on neuroblastoma with MAPK pathway mutations. As we observed eventual resistance to trametinib in our previous study, we evaluated the combination therapy of CA3, a YAP inhibitor, with trametinib, based on a recent report suggesting the potential involvement of YAP in the mechanism underlying the resistance to trametinib in neuroblastoma.. SK-N-AS cells (a neuroblastoma cell line harboring RAS mutation) were treated with CA3 in vitro and subjected to a viability assay, immunocytochemistry and flow cytometry. Next, we analyzed the in vitro combination effect of CA3 and trametinib using the CompuSyn software program. Finally, we administered CA3, trametinib or both to SK-N-AS xenograft mice for 10 weeks to analyze the combination effect.. CA3 inhibited cell proliferation by both cell cycle arrest and apoptosis in vitro. Combination of CA3 and trametinib induced a significant synergistic effect in vitro (Combination Index <1). Regarding the in vivo experiment, combination therapy suppressed tumor growth, and 100% of mice in the combination therapy group survived, whereas the survival rates were 0% in the CA3 group and 33% in the trametinib group. However, despite this promising survival rate in the combination group, the tumors gradually grew after seven weeks with MAPK reactivation.. Our results indicated that CA3 and trametinib exerted synergistic antitumor effects on neuroblastoma in vitro and in vivo, and CA3 may be a viable option for concomitant drug therapy with trametinib, since it suppressed the resistance to trametinib. However, this combination effect was not sufficient to achieve complete remission. Therefore, we need to adjust the protocol to obtain a better outcome by determining the mechanism underlying regrowth in the future. Topics: Adaptor Proteins, Signal Transducing; Animals; Antineoplastic Combined Chemotherapy Protocols; Apoptosis; Cell Line, Tumor; Cell Proliferation; Drug Synergism; Extracellular Signal-Regulated MAP Kinases; Female; Mice, Nude; Mitogen-Activated Protein Kinase Kinases; Neuroblastoma; Phosphorylation; Protein Kinase Inhibitors; Pyridones; Pyrimidinones; S Phase; Survival Analysis; Transcription Factors; Xenograft Model Antitumor Assays; YAP-Signaling Proteins | 2021 |
Inhibition of MEK pathway enhances the antitumor efficacy of chimeric antigen receptor T cells against neuroblastoma.
Disialoganglioside (GD2)-specific chimeric antigen receptor (CAR)-T cells (GD2-CAR-T cells) have been developed and tested in early clinical trials in patients with relapsed/refractory neuroblastoma. However, the effectiveness of immunotherapy using these cells is limited, and requires improvement. Combined therapy with CAR-T cells and molecular targeted drugs could be a promising strategy to enhance the antitumor efficacy of CAR T cell immunotherapy. Here, we generated GD2-CAR-T cells through piggyBac transposon (PB)-based gene transfer (PB-GD2-CAR-T cells), and analyzed the combined effect of these cells and a MEK inhibitor in vitro and in vivo on neuroblastoma. Trametinib, a MEK inhibitor, ameliorated the killing efficacy of PB-GD2-CAR-T cells in vitro, whereas a combined treatment of the two showed superior antitumor efficacy in a murine xenograft model compared to that of PB-GD2-CAR-T cell monotherapy, regardless of the mutation status of the MAPK pathway in tumor cells. The results presented here provide new insights into the feasibility of combined treatment with CAR-T cells and MEK inhibitors in patients with neuroblastoma. Topics: Animals; Antineoplastic Agents; Cell Line, Tumor; Combined Modality Therapy; Coumarins; DNA Transposable Elements; Drug Resistance, Neoplasm; Female; Gangliosides; Genetic Therapy; Humans; Immunotherapy, Adoptive; Mice; Mice, SCID; Mitogen-Activated Protein Kinase Kinases; Mutation; Neoplasm Recurrence, Local; Neuroblastoma; Protein Kinase Inhibitors; Pyridones; Pyrimidinones; ras Proteins; Receptors, Chimeric Antigen; T-Lymphocytes; Xenograft Model Antitumor Assays | 2021 |
Preclinical evaluation of the combination of AZD1775 and irinotecan against selected pediatric solid tumors: A Pediatric Preclinical Testing Consortium report.
WEE1 is a serine kinase central to the G. AZD1775 was tested using a dose of 120 mg/kg administered orally for days 1 to 5. Irinotecan was administered intraperitoneally at a dose of 2.5 mg/kg for days 1 to 5 (one hour after AZD1775 when used in combination). AZD1775 and irinotecan were studied alone and in combination in neuroblastoma (n = 3), osteosarcoma (n = 4), and Wilms tumor (n = 3) xenografts.. AZD1775 as a single agent showed little activity. Irinotecan induced objective responses in two neuroblastoma lines (PRs), and two Wilms tumor models (CR and PR). The combination of AZD1775 + irinotecan-induced objective responses in two neuroblastoma lines (PR and CR) and all three Wilms tumor lines (CR and 2 PRs). The objective response measure improved compared with single-agent treatment for one neuroblastoma (PR to CR), two osteosarcoma (PD1 to PD2), and one Wilms tumor (PD2 to PR) xenograft lines. Of note, the combination yielded CR (n = 1) and PR (n = 2) in all the Wilms tumor lines. The event-free survival was significantly longer for the combination compared with single-agent irinotecan in all models tested. The magnitude of the increase was greatest in osteosarcoma and Wilms tumor xenografts.. AZD1775 potentiates the effects of irinotecan across most of the xenograft lines tested, with effect size appearing to vary across tumor panels. Topics: Animals; Antineoplastic Combined Chemotherapy Protocols; Cell Line, Tumor; Child; Female; Humans; Irinotecan; Kidney Neoplasms; Mice; Mice, SCID; Neoplasms, Experimental; Neuroblastoma; Pyrazoles; Pyrimidinones; Wilms Tumor; Xenograft Model Antitumor Assays | 2020 |
Limited antitumor activity of combined BET and MEK inhibition in neuroblastoma.
The treatment of high-risk neuroblastoma continues to present a formidable challenge to pediatric oncology. Previous studies have shown that Bromodomain and extraterminal (BET) inhibitors can inhibit MYCN expression and suppress MYCN-amplified neuroblastoma in vivo. Furthermore, alterations within RAS-MAPK (mitogen-activated protein kinase) signaling play significant roles in neuroblastoma initiation, maintenance, and relapse, and mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors demonstrate efficacy in subsets of neuroblastoma preclinical models. Finally, hyperactivation of RAS-MAPK signaling has been shown to promote resistance to BET inhibitors. Therefore, we examined the antitumor efficacy of combined BET/MEK inhibition utilizing I-BET726 or I-BET762 and trametinib in high-risk neuroblastoma.. Utilizing a panel of genomically annotated neuroblastoma cell line models, we investigated the in vitro effects of combined BET/MEK inhibition on cell proliferation and apoptosis. Furthermore, we evaluated the effects of combined inhibition in neuroblastoma xenograft models.. Combined BET and MEK inhibition demonstrated synergistic effects on the growth and survival of a large panel of neuroblastoma cell lines through augmentation of apoptosis. A combination therapy slowed tumor growth in a non-MYCN-amplified, NRAS-mutated neuroblastoma xenograft model, but had no efficacy in an MYCN-amplified model harboring a loss-of-function mutation in NF1.. Combinatorial BET and MEK inhibition was synergistic in the vast majority of neuroblastoma cell lines in the in vitro setting but showed limited antitumor activity in vivo. Collectively, these data do not support clinical development of this combination in high-risk neuroblastoma. Topics: Animals; Antineoplastic Agents; Apoptosis; Benzodiazepines; Cell Proliferation; Female; Humans; MAP Kinase Kinase 1; Mice; Mice, SCID; Neuroblastoma; Proteins; Pyridones; Pyrimidinones; Tumor Cells, Cultured; Xenograft Model Antitumor Assays | 2020 |
Survival for high-risk neuroblastoma remains poor and treatment for relapsed disease rarely leads to long-term cures. Large sequencing studies of neuroblastoma tumors from diagnosis have not identified common targetable driver mutations other than the 10% of tumors that harbor mutations in the anaplastic lymphoma kinase ( Topics: Aminopyridines; Animals; Antineoplastic Agents; Cell Line, Tumor; Drug Resistance, Neoplasm; Drug Synergism; Genes, ras; Heterografts; Humans; Indoles; Male; MAP Kinase Signaling System; Mice, Inbred NOD; Mice, SCID; Mitogen-Activated Protein Kinase Kinases; Mutation; Neoplasm Recurrence, Local; Neuroblastoma; Piperidines; Protein Tyrosine Phosphatase, Non-Receptor Type 11; Pyridones; Pyrimidines; Pyrimidinones; Pyrroles; Triazoles; Vemurafenib | 2020 |
YAP1 Mediates Resistance to MEK1/2 Inhibition in Neuroblastomas with Hyperactivated RAS Signaling.
Topics: Adaptor Proteins, Signal Transducing; Cell Line, Tumor; Drug Resistance, Neoplasm; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; MAP Kinase Signaling System; Mutation; Neuroblastoma; Protein Kinase Inhibitors; Proto-Oncogene Proteins p21(ras); Pyridones; Pyrimidinones; Transcription Factors; YAP-Signaling Proteins | 2019 |
Prohibitin promotes de-differentiation and is a potential therapeutic target in neuroblastoma.
Gain of the long arm of chromosome 17 (17q) is a cytogenetic hallmark of high-risk neuroblastoma, yet its contribution to neuroblastoma pathogenesis remains incompletely understood. Combining whole-genome and RNA sequencing of neuroblastomas, we identified the prohibitin (PHB) gene as highly expressed in tumors with 17q gain. High PHB expression correlated with poor prognosis and was associated with loss of gene expression programs promoting neuronal development and differentiation. PHB depletion induced differentiation and apoptosis and slowed cell cycle progression of neuroblastoma cells, at least in part through impaired ERK1/2 activation. Conversely, ectopic expression of PHB was sufficient to increase proliferation of neuroblastoma cells and was associated with suppression of markers associated with neuronal differentiation and favorable neuroblastoma outcome. Thus, PHB is a 17q oncogene in neuroblastoma that promotes tumor cell proliferation, and de-differentiation. Topics: Animals; Apoptosis; Cell Cycle Checkpoints; Cell Dedifferentiation; Cell Differentiation; Cell Line, Tumor; Cell Proliferation; Child, Preschool; Chromosomes, Human, Pair 17; Humans; MAP Kinase Signaling System; Mice; Neuroblastoma; Prohibitins; Protein Kinase Inhibitors; Pyridones; Pyrimidinones; Repressor Proteins; RNA-Seq; RNA, Messenger; Sequence Analysis, RNA; Whole Genome Sequencing; Xenograft Model Antitumor Assays | 2019 |
MRE11 inhibition highlights a replication stress-dependent vulnerability of MYCN-driven tumors.
MRE11 is a component of the MRE11/RAD50/NBS1 (MRN) complex, whose activity is essential to control faithful DNA replication and to prevent accumulation of deleterious DNA double-strand breaks. In humans, hypomorphic mutations in these genes lead to DNA damage response (DDR)-defective and cancer-prone syndromes. Moreover, MRN complex dysfunction dramatically affects the nervous system, where MRE11 is required to restrain MYCN-dependent replication stress, during the rapid expansion of progenitor cells. MYCN activation, often due to genetic amplification, represents the driving oncogenic event for a number of human tumors, conferring bad prognosis and predicting very poor responses even to the most aggressive therapeutic protocols. This is prototypically exemplified by neuroblastoma, where MYCN amplification occurs in about 25% of the cases. Intriguingly, MRE11 is highly expressed and predicts bad prognosis in MYCN-amplified neuroblastoma. Due to the lack of direct means to target MYCN, we explored the possibility to trigger intolerable levels of replication stress-dependent DNA damage, by inhibiting MRE11 in MYCN-amplified preclinical models. Indeed, either MRE11 knockdown or its pharmacological inhibitor mirin induce accumulation of replication stress and DNA damage biomarkers in MYCN-amplified cells. The consequent DDR recruits p53 and promotes a p53-dependent cell death, as indicated by p53 loss- and gain-of-function experiments. Encapsulation of mirin in nanoparticles allowed its use on MYCN-amplified neuroblastoma xenografts in vivo, which resulted in a sharp impairment of tumor growth, associated with DDR activation, p53 accumulation, and cell death. Therefore, we propose that MRE11 inhibition might be an effective strategy to treat MYCN-amplified and p53 wild-type neuroblastoma, and suggest that targeting replication stress with appropriate tools should be further exploited to tackle MYCN-driven tumors. Topics: 3T3 Cells; A549 Cells; Animals; Apoptosis; Cell Line, Tumor; Cell Proliferation; Cell Survival; DNA Damage; Female; HEK293 Cells; Hep G2 Cells; Humans; Mice; Mice, Inbred BALB C; Mice, Nude; MRE11 Homologue Protein; N-Myc Proto-Oncogene Protein; Neuroblastoma; Prognosis; Pyrimidinones; Thiones; Tumor Suppressor Protein p53; Xenograft Model Antitumor Assays | 2018 |
In vivo effects of short- and long-term MAPK pathway inhibition against neuroblastoma.
It was reported that almost 80% of relapsed neuroblastomas showed MAPK pathway mutations. In our previous study, both trametinib (MEK inhibitor) and CH5126766 (RAF/MEK inhibitor) showed in vitro antitumor effects on neuroblastoma cells with ERK phosphorylation (pERK). In this study, we analyzed the in vivo effects of MAPK pathway inhibition in neuroblastoma xenografts.. Xenograft mice with IMR5, CHP-212, or SK-N-AS received daily oral administration of either trametinib or CH5126766 for two weeks (short term) or eight weeks (long term). The tumors were measured twice weekly and harvested after treatment for histopathological analyses, including pERK and Ki67 immunohistochemistry.. In short-term treatment, both inhibitors showed significant growth inhibition in CHP-212 and SK-N-AS xenografts, which were pERK-positive before treatment. The number of pERK- and Ki67-positive cells decreased after treatment. Conversely, IMR5 xenografts, which were pERK-negative, were resistant to treatment. During long-term treatment, SK-N-AS xenografts started to regrow from about six weeks with partial differentiation. pERK-positive cells reincreased in these regrown tumors.. MAPK pathway inhibition was effective for treating pERK-positive neuroblastoma in vivo. Therefore, pERK immunohistochemistry might be a convenient biomarker for MAPK pathway inhibition in neuroblastoma treatment. However, neuroblastomas developed acquired drug resistance after long-term treatment. Further studies to overcome acquired resistance are needed. Topics: Animals; Cell Line, Tumor; Coumarins; eIF-2 Kinase; Female; Humans; Immunohistochemistry; Ki-67 Antigen; Mice; Mitogen-Activated Protein Kinases; Neoplasm Recurrence, Local; Neuroblastoma; Protein Kinase Inhibitors; Pyridones; Pyrimidinones; Xenograft Model Antitumor Assays | 2018 |
MEK inhibitor trametinib does not prevent the growth of anaplastic lymphoma kinase (ALK)-addicted neuroblastomas.
Activation of the RAS-RAF-MEK-ERK signaling pathway is implicated in driving the initiation and progression of multiple cancers. Several inhibitors targeting the RAS-MAPK pathway are clinically approved as single- or polyagent therapies for patients with specific types of cancer. One example is the MEK inhibitor trametinib, which is included as a rational polytherapy strategy for treating EML4-ALK-positive, EGFR-activated, or KRAS-mutant lung cancers and neuroblastomas that also contain activating mutations in the RAS-MAPK pathway. In addition, in neuroblastoma, a heterogeneous disease, relapse cases display an increased rate of mutations in Topics: Anaplastic Lymphoma Kinase; Animals; Antineoplastic Agents; Cell Line, Tumor; Female; Humans; Lung Neoplasms; MAP Kinase Signaling System; Mechanistic Target of Rapamycin Complex 2; Mice; Mice, Inbred BALB C; Mitogen-Activated Protein Kinase 7; Neuroblastoma; Oncogene Proteins, Fusion; Protein Kinase Inhibitors; Proto-Oncogene Proteins c-akt; Pyridones; Pyrimidinones; Receptor Protein-Tyrosine Kinases; Xenograft Model Antitumor Assays | 2017 |
Wnt signalling is a bi-directional vulnerability of cancer cells.
Wnt signalling is involved in the formation, metastasis and relapse of a wide array of cancers. However, there is ongoing debate as to whether activation or inhibition of the pathway holds the most promise as a therapeutic treatment for cancer, with conflicting evidence from a variety of tumour types. We show that Wnt/β-catenin signalling is a bi-directional vulnerability of neuroblastoma, malignant melanoma and colorectal cancer, with hyper-activation or repression of the pathway both representing a promising therapeutic strategy, even within the same cancer type. Hyper-activation directs cancer cells to undergo apoptosis, even in cells oncogenically driven by β-catenin. Wnt inhibition blocks proliferation of cancer cells and promotes neuroblastoma differentiation. Wnt and retinoic acid co-treatments synergise, representing a promising combination treatment for MYCN-amplified neuroblastoma. Additionally, we report novel cross-talks between MYCN and β-catenin signalling, which repress normal β-catenin mediated transcriptional regulation. A β-catenin target gene signature could predict patient outcome, as could the expression level of its DNA binding partners, the TCF/LEFs. This β-catenin signature provides a tool to identify neuroblastoma patients likely to benefit from Wnt-directed therapy. Taken together, we show that Wnt/β-catenin signalling is a bi-directional vulnerability of a number of cancer entities, and potentially a more broadly conserved feature of malignant cells. Topics: Antineoplastic Agents; beta Catenin; Bridged Bicyclo Compounds, Heterocyclic; Cell Differentiation; Cell Line, Tumor; Cell Proliferation; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Humans; N-Myc Proto-Oncogene Protein; Neuroblastoma; Proteomics; Pyrimidinones; RNA Interference; Survival Analysis; Tretinoin; Wnt Proteins; Wnt Signaling Pathway | 2016 |
MEK inhibitors as a novel therapy for neuroblastoma: Their in vitro effects and predicting their efficacy.
A recent study reported that relapsed neuroblastomas had frequent RAS-ERK pathway mutations. We herein investigated the effects and pathways of MEK inhibitors, which inhibit the RAS-ERK pathway, as a new molecular-targeted therapy for refractory neuroblastomas.. Five neuroblastoma cell lines were treated with trametinib (MEK inhibitor) or CH5126766 (RAF/MEK inhibitor). Growth inhibition was analyzed using a cell viability assay. ERK phosphorylation and the MYCN expression were analyzed by immunoblotting or immunohistochemistry. RAS/RAF mutations were identified by direct sequencing or through the COSMIC database.. Both MEK inhibitors showed growth inhibition effects on cells with ERK phosphorylation, but almost no effect on cells without. In immunoblotting analyses, ERK phosphorylation and MYCN expression were suppressed in ERK active cells by these drugs. Furthermore, phosphorylated-ERK immunohistochemistry corresponded to the drug responses. Regarding the relationship between RAS/Raf mutations and ERK phosphorylation, ERK was phosphorylated in one cell line (NLF) without RAS/Raf mutations.. MEK inhibitors are a promising molecular-targeted therapeutic option for ERK active neuroblastomas. The efficacy of MEK inhibitors corresponds to ERK phosphorylation, while RAS/RAF mutations are not always detected in drug-sensitive cells. Phosphorylated-ERK immunohistochemistry is thus a useful method to analyze ERK activity and predict the therapeutic effects of MEK inhibitors. Topics: Cell Line, Tumor; Coumarins; Humans; MAP Kinase Signaling System; Molecular Targeted Therapy; Neoplasm Recurrence, Local; Neuroblastoma; Phosphorylation; Protein Kinase Inhibitors; Pyridones; Pyrimidinones; Signal Transduction | 2016 |
Stress Conditions Increase Vimentin Cleavage by Omi/HtrA2 Protease in Human Primary Neurons and Differentiated Neuroblastoma Cells.
Dysfunctional Omi/HtrA2, a mitochondrial serine protease, has been implicated in various neurodegenerative disorders. Despite the wealth of evidence on the roles of Omi/HtrA2 in apoptosis, little is known about its cytosolic targets, the cleavage of which could account for the observed morphological changes such as cytoskeletal reorganizations in axons. By proteomic analysis, vimentin was identified as a substrate for Omi/HtrA2 and we have reported increased Omi/HtrA2 protease activity in Alzheimer disease (AD) brain. Here, we investigated a possible link between Omi/HtrA2 and vimentin cleavage, and consequence of this cleavage on mitochondrial distribution in neurons. In vitro protease assays showed vimentin to be cleaved by Omi/HtrA2 protease, and proximity ligation assay demonstrated an increased interaction between Omi/HtrA2 and vimentin in human primary neurons upon stress stimuli. Using differentiated neuroblastoma SH-SY5Y cells, we showed that Omi/HtrA2 under several different stress conditions induces cleavage of vimentin in wild-type as well as SH-SY5Y cells transfected with amyloid precursor protein with the Alzheimer disease-associated Swedish mutation. After stress treatment, inhibition of Omi/HtrA2 protease activity by the Omi/HtrA2 specific inhibitor, Ucf-101, reduced the cleavage of vimentin in wild-type cells. Following altered vimentin filaments integrity by stress stimuli, mitochondria was redistributed in differentiated SH-SY5Y cells and human primary neurons. In summary, the findings outlined in this paper suggest a role of Omi/HtrA2 in modulation of vimentin filamentous structure in neurons. Our results provide important findings for understanding the biological role of Omi/HtrA2 activity during stress conditions, and give knowledge of interplay between Omi/HtrA2 and vimentin which might affect mitochondrial distribution in neurons. Topics: Amyloid beta-Protein Precursor; Axons; Cell Line, Tumor; Culture Media, Serum-Free; Cytosol; High-Temperature Requirement A Serine Peptidase 2; Humans; Interleukin-1beta; Intermediate Filaments; Isothiocyanates; Mitochondria; Mitochondrial Proteins; Mutation, Missense; Neoplasm Proteins; Nerve Tissue Proteins; Neuroblastoma; Neurogenesis; Neurons; Point Mutation; Pyrimidinones; Serine Endopeptidases; Serine Proteinase Inhibitors; Stress, Physiological; Sulfoxides; Thiones; Transfection; Tretinoin; Tunicamycin; Vimentin | 2015 |
Targeting histone deacetylases (HDACs) and Wee1 for treating high-risk neuroblastoma.
Despite advances in treatment regimens, patients with high-risk neuroblastoma have long-term survival rates of < 40%. Wee1 inhibition in combination with CHK1 inhibition has shown promising results in neuroblastoma cells. In addition, it has been demonstrated that panobinostat can downregulate CHK1. Therefore, combination of panobinostat and MK-1775 may result in synergistic cytotoxicity against neuroblastoma cell lines.. In vitro cytotoxicities of panobinostat and MK-1775 at clinically achievable concentrations, either alone or in combination, were evaluated in SK-N-AS, SK-N-DZ, and SK-N-BE(2) high-risk neuroblastoma cell lines using MTT assays. The mechanism of antitumor interaction was investigated using propidium iodide (PI) staining and flow cytometry analysis to determine apoptosis, as well as Western blotting to assess expression of phosphorylated CDK1/2, CHK1, and H2AX.. Treatment of neuroblastoma cell lines with 500 nM MK-1775 caused growth arrest and apoptosis in SK-N-DZ and SK-N-AS, while it had minimal effect on the SK-N-BE(2) cell line. The combination of panobinostat and MK-1775 resulted in synergistic antitumor interactions in all three of the cell lines tested. MK-1775 treatment in SK-N-BE(2) cells induced increased levels of p-CHK1(S345) , which could be decreased by the addition of panobinostat. This was accompanied by increased DNA damage and apoptosis.. The combination of panobinostat and MK-1775 has synergistic antitumor activity against neuroblastoma cell lines and holds promise as a potential treatment strategy for the management of high-risk neuroblastoma patients. Topics: Apoptosis; Blotting, Western; Cell Cycle; Cell Cycle Proteins; Cell Proliferation; Drug Synergism; Drug Therapy, Combination; Histone Deacetylase Inhibitors; Histone Deacetylases; Humans; Hydroxamic Acids; Indoles; Neuroblastoma; Nuclear Proteins; Panobinostat; Protein-Tyrosine Kinases; Pyrazoles; Pyrimidines; Pyrimidinones; Risk Factors; Tumor Cells, Cultured | 2015 |
Trametinib-induced Left Ventricular Dysfunction in a Child With Relapsed Neuroblastoma.
The MEK inhibitor trametinib is globally approved for metastatic melanoma harboring BRAF mutations. There are no reports thus far on its use in children. Exome sequencing on a relapsed tumor sample from an 11-year-old male with progressive, multiply relapsed stage 4 neuroblastoma revealed NRASQ61K mutation. After demonstration of normal cardiac function, he was started on oral trametinib. On day 13 of treatment, echocardiogram showed moderate left ventricular dysfunction. Trametinib was discontinued on day 15 and oral lisinopril was started. Left ventricular function recovered to baseline 37 days after discontinuing trametinib. However, neuroblastoma showed further progression. Topics: Child; GTP Phosphohydrolases; Humans; Male; Membrane Proteins; Mutation; Neoplasm Recurrence, Local; Neuroblastoma; Prognosis; Protein Kinase Inhibitors; Pyridones; Pyrimidinones; Ventricular Dysfunction, Left | 2015 |
Combination therapy targeting the Chk1 and Wee1 kinases shows therapeutic efficacy in neuroblastoma.
Neuroblastoma is uniquely sensitive to single-agent inhibition of the DNA damage checkpoint kinase Chk1, leading us to examine downstream effectors of this pathway and identify mitotic regulator Wee1 as an additional therapeutic target in this disease. Wee1 was overexpressed in both neuroblastoma cell lines and high-risk patient tumors. Genetic or pharmacologic abrogation of Wee1 signaling results in marked cytotoxicity in 10 of 11 neuroblastoma cell lines with a median IC(50) of 300 nmol/L for the Wee1-selective small-molecule inhibitor MK-1775. Murine tumor lines derived from mice that were either heterozygous or homozygous for MycN were particularly sensitive to single-agent inhibition of Wee1 (IC(50)s of 160 and 62 nmol/L, respectively). Simultaneous pharmacologic inhibition of Chk1 and Wee1 acted in a synergistic fashion to further impede neuroblastoma cell growth in vitro, in a manner greater than the individual inhibitors either alone or combined with chemotherapy. Combination Chk1 and Wee1 inhibition also revealed in vivo efficacy in neuroblastoma xenografts. Taken together, our results show that neuroblastoma cells depend on Wee1 activity for growth and that inhibition of this kinase may serve as a therapeutic for patients with neuroblastoma. Topics: Animals; Antineoplastic Combined Chemotherapy Protocols; Cell Cycle Proteins; Cell Line, Tumor; Checkpoint Kinase 1; Female; Humans; Mice; Mice, SCID; Neuroblastoma; Nuclear Proteins; Protein Kinase Inhibitors; Protein Kinases; Protein-Tyrosine Kinases; Pyrazoles; Pyrimidines; Pyrimidinones; Xenograft Model Antitumor Assays | 2013 |
Efficacy of class I and II vs class III histone deacetylase inhibitors in neuroblastoma.
Histone deacetylase (HDAC) inhibitors have shown promise in the treatment of resistant and refractory tumors including neuroblastoma. The goal of the study was to compare the efficacy of a class III HDAC inhibitor (cambinol) to a class I and II inhibitor (vorinostat).. In vitro efficacy of vorinostat and cambinol, alone or in combination with doxorubicin, was assessed by 2-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide calorimetric assay using both wild-type (WT) and doxorubicin-resistant (DoxR) SK-N-SH neuroblastoma cells. In vivo efficacy was determined using the same drug combinations in nude mice bearing xenograft implants of WT and DoxR cells on opposite flanks.. Vorinostat and cambinol were efficacious against WT and DoxR neuroblastoma cells in vitro. In WT cells, the potency of the doxorubicin itself overshadowed any effect of cotherapy with vorinostat or cambinol. The effect of vorinostat and/or cambinol on the DoxR cells was constant across progressively increasing doses of doxorubicin. In the in vivo model, the efficacy of doxorubicin itself (88% reduction in tumor volume) again overshadowed any effect of cotreatment with vorinostat or cambinol on the WT tumors. However, in the DoxR tumors, doxorubicin alone had no efficacy, but cotreatment with either cambinol or vorinostat suppressed tumor growth (70% and 91% reduction in tumor volume, respectively).. Both the class III HDAC inhibitor cambinol and the class I/II HDAC inhibitor vorinostat have efficacy against SK-N-SH neuroblastoma cells, including those resistant to doxorubicin. Topics: Animals; Antibiotics, Antineoplastic; Antineoplastic Agents; Antineoplastic Combined Chemotherapy Protocols; Cell Line, Tumor; Doxorubicin; Drug Resistance, Neoplasm; Drug Synergism; Histone Deacetylase Inhibitors; Humans; Hydroxamic Acids; Mice; Mice, Nude; Naphthalenes; Neoplasm Proteins; Neuroblastoma; Pyrimidinones; Tumor Stem Cell Assay; Vorinostat; Xenograft Model Antitumor Assays | 2012 |
Wnt pathway activity confers chemoresistance to cancer stem-like cells in a neuroblastoma cell line.
Neuroblastoma is the most common solid tumor in infancy. We have shown that the neuroblastoma cell line SK-N-SH contains CD133+ cells that are more resistant than 133- cells to Doxorubicin (DOX), a common chemotherapeutic agent. We hypothesize that activation of wnt signaling pathway in CD133+ cells contributes to their chemoresistance. To test this hypothesis, CD133+ cells were positively selected using magnetic micro-beads. Subsequently, CD133+ and negatively selected CD133- cells were treated with 100 ng/ml of DOX for up to 72 h. Then, cells were either lysed for total RNA extraction or fixed for immunostaining. Wnt "SIGNATURE" PCR Array was used to determine if changes in wnt related gene expression levels occurred and to estimate a pathway activity score. Expression of wnt pathway proteins β-Catenin and p-GSK3β (S-9) was determined by immunocytochemistry. Two wnt pathway inhibitors were used to determine the changes in cell viability, using the MTT assay. Results showed that wnt related genes were differentially expressed in CD133+ cells as compared to CD133- cells, both with and without DOX treatment. Pathway activity scores showed that DOX treatment significantly suppressed the wnt pathway activity in CD133- cells. Expression of β-catenin and p-GSK3β (S-9) was significantly greater in DOX treated and untreated CD133+ cells. The presence of wnt inhibitors with DOX decreased the number of live cells in CD133+ group and the percentage of live cells in both groups were equal. These data suggest that higher wnt pathway activity could be responsible for the chemoresistance of CD133+ cells in neuroblastoma cell lines. Topics: AC133 Antigen; Antibiotics, Antineoplastic; Antigens, CD; beta Catenin; Blotting, Western; Bridged Bicyclo Compounds, Heterocyclic; Cell Proliferation; Doxorubicin; Drug Resistance, Neoplasm; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Glycoproteins; Heterocyclic Compounds, 3-Ring; Humans; Immunoenzyme Techniques; Immunophenotyping; Neoplastic Stem Cells; Neuroblastoma; Peptides; Pyrimidinones; Real-Time Polymerase Chain Reaction; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Tumor Cells, Cultured; Wnt Proteins; Wnt Signaling Pathway | 2012 |
SIRT1 promotes N-Myc oncogenesis through a positive feedback loop involving the effects of MKP3 and ERK on N-Myc protein stability.
The N-Myc oncoprotein is a critical factor in neuroblastoma tumorigenesis which requires additional mechanisms converting a low-level to a high-level N-Myc expression. N-Myc protein is stabilized when phosphorylated at Serine 62 by phosphorylated ERK protein. Here we describe a novel positive feedback loop whereby N-Myc directly induced the transcription of the class III histone deacetylase SIRT1, which in turn increased N-Myc protein stability. SIRT1 binds to Myc Box I domain of N-Myc protein to form a novel transcriptional repressor complex at gene promoter of mitogen-activated protein kinase phosphatase 3 (MKP3), leading to transcriptional repression of MKP3, ERK protein phosphorylation, N-Myc protein phosphorylation at Serine 62, and N-Myc protein stabilization. Importantly, SIRT1 was up-regulated, MKP3 down-regulated, in pre-cancerous cells, and preventative treatment with the SIRT1 inhibitor Cambinol reduced tumorigenesis in TH-MYCN transgenic mice. Our data demonstrate the important roles of SIRT1 in N-Myc oncogenesis and SIRT1 inhibitors in the prevention and therapy of N-Myc-induced neuroblastoma. Topics: Animals; Binding Sites; Cell Line, Tumor; Cell Proliferation; Dual Specificity Phosphatase 6; Enzyme Inhibitors; Extracellular Signal-Regulated MAP Kinases; Feedback, Physiological; Gene Expression Regulation, Neoplastic; Mice; Mice, Transgenic; Naphthalenes; Neuroblastoma; Phosphorylation; Promoter Regions, Genetic; Protein Stability; Proto-Oncogene Proteins c-myc; Pyrimidinones; Random Allocation; Sirtuin 1; Sp1 Transcription Factor; Tumor Burden | 2011 |
The role of zinc in the modulation of neuronal proliferation and apoptosis.
Although a requirement of zinc (Zn) for normal brain development is well documented, the extent to which Zn can modulate neuronal proliferation and apoptosis is not clear. Thus, we investigated the role of Zn in the regulation of these two critical events. A low Zn availability leads to decreased cell viability in human neuroblastoma IMR-32 cells and primary cultures of rat cortical neurons. This occurs in part as a consequence of decreased cell proliferation and increased apoptotic cell death. In IMR-32 cells, Zn deficiency led to the inhibition of cell proliferation through the arrest of the cell cycle at the G0/G1 phase. Zn deficiency induced apoptosis in both proliferating and quiescent neuronal cells via the intrinsic apoptotic pathway. Reductions in cellular Zn triggered a translocation of the pro-apoptotic protein Bad to the mitochondria, cytochrome c release, and caspase-3 activation. Apoptosis is the resultant of the inhibition of the prosurvival extracellular-signal-regulated kinase, the inhibition of nuclear factor-kappa B, and associated decreased expression of antiapoptotic proteins, and to a direct activation of caspase-3. A deficit of Zn during critical developmental periods can have persistent effects on brain function secondary to a deregulation of neuronal proliferation and apoptosis. Topics: Analysis of Variance; Animals; Antioxidants; Apoptosis; Caspase 3; Cell Cycle; Cell Proliferation; Cell Survival; Cells, Cultured; Cerebral Cortex; Cytochromes c; Dose-Response Relationship, Drug; Electrophoretic Mobility Shift Assay; Embryo, Mammalian; Female; Gene Expression Regulation; Humans; In Situ Nick-End Labeling; Mitogen-Activated Protein Kinases; Neuroblastoma; Neurons; Photosensitizing Agents; Pregnancy; Proto-Oncogene Proteins c-bcl-2; Pyrimidinones; Rats; Rats, Sprague-Dawley; Serine; Signal Transduction; Thioctic Acid; Time Factors; Zinc | 2010 |
Preferential inactivation of paediatric solid tumour cells by sequential exposure to Merocyanine 540-mediated photodynamic therapy and Edelfosine: implications for the ex vivo purging of autologous haematopoietic stem cell grafts.
Paediatric solid tumours exhibit steep dose-response curves to alkylating agents and are therefore considered candidates for high-dose chemotherapy and autologous stem cell support. There is growing evidence that autologous stem cell grafts from patients with solid tumours are frequently contaminated with live tumour cells. The objective of this study was to perform, in a preclinical purging model, an initial assessment of the safety and efficacy of a two-step purging procedure that combined Merocyanine 540-mediated photodynamic therapy (MC540-PDT) with a brief exposure to the alkyl-lysophospholipid, Edelfosine. Human and murine bone marrow cells and Neuro-2a murine neuroblastoma, SK-N-SH human neuroblastoma, SK-ES-1 and U-2 OS human osteosarcoma, G-401 and SK-NEP-1 human Wilms' tumour, and A-204 human rhabdomyosarcoma cells were exposed to a fixed dose of MC540-PDT followed by a brief incubation with graded concentrations of Edelfosine. Survival was subsequently assessed by in vitro clonal assay or, in the case of CD34-positive haematopoietic stem cells, by an immunohistochemical method. Combination purging with MC540-PDT and Edelfosine depleted all tumour cells by >4 log while preserving at least 15% of murine granulocyte/macrophage progenitors (CFU-GM), 34% of human CFU-GM, and 31% of human CD34-positive cells. The data suggest that combination purging with MC540-PDT and Edelfosine may be useful for the ex vivo purging of autologous stem cell grafts from patients with paediatric solid tumours. Topics: Animals; Antineoplastic Combined Chemotherapy Protocols; Cell Survival; Dose-Response Relationship, Drug; Hematopoietic Stem Cell Transplantation; Humans; Mice; Neuroblastoma; Osteosarcoma; Phospholipid Ethers; Photochemotherapy; Photosensitizing Agents; Pyrimidinones; Rhabdomyosarcoma; Tumor Cells, Cultured; Wilms Tumor | 2003 |
Merocyanine 540 mediated photolysis of normal bone marrow, committed hemopoietic progenitors and neoplastic cells. implications for bone marrow purging.
The effect of merocyanine 540 (Mc 540) mediated photoirradiation on both neoplastic and normal hemopoietic progenitor cells was studied. Bone marrow (BM) cells from children with acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML) at initial diagnosis, ALL in remission, neuroblastoma and normal children as well as cells of Reh-6 and HL-60 cell lines were incubated with Mc 540 in the presence of human albumin (HA) and exposed to different argon laser 514 nm doses. Cell survival was estimated using Trypan Blue supravital stain following a 24-h incubation and leukemic cell lines were studied in continuous cell cultures of 4 weeks duration. Our results showed that HA protects normal BM cells from Mc 540 mediated phototoxicity. A 99.9999% inhibition of Reh-6 and HL-60 was noted at irradiation doses where the corresponding mean survival of normal BM cells was 77.4 +/- 12 and 70.3 +/- 10%, respectively. BM leukemic cells from children with ALL and AML were also very sensitive to Mc 540 photoirradiation in contrast to neuroblastoma cells where only a three-fold reduction was observed. Finally, the survival of normal BM progenitors was 38% for colony forming unit erythroid CFU-E, 37% for burst forming unit erythroid BFU-E, 55% for CFU-GM and 29% for CFU-GEMM. In conclusion it seems that Mc 540 mediated photoirradiation in neoplastic cells exerts selective cytotoxicity and can be used in ex vivo purging of malignant cells in BM. Topics: Bone Marrow; Bone Marrow Cells; Bone Marrow Purging; Cell Line; Cell Survival; Child; Dose-Response Relationship, Radiation; Hematopoietic Stem Cells; HL-60 Cells; Humans; Lasers; Leukemia, Myeloid, Acute; Neuroblastoma; Photolysis; Precursor Cell Lymphoblastic Leukemia-Lymphoma; Pyrimidinones; Radiation-Sensitizing Agents | 1997 |
Mechanism of photosensitization of glioblastoma and neuroblastoma cells by merocyanine 540: a lipid peroxidation study.
Mechanism of merocyanine 540 (MC540) mediated photosensitization in glioblastoma (U-87MG) and neuroblastoma (Neuro 2a) cells was investigated. Photoinduced lipid peroxidation was measured in the presence of mechanistic probes-deuterium oxide (D2O), sodium azide, superoxide dismutase (SOD), mannitol and sodium benzoate. In both the types of cells, the photoinduced lipid peroxidation was enhanced in D2O whereas it showed inhibition in the presence of sodium azide. SOD also inhibited the lipid peroxidation while sodium benzoate and mannitol had no effect. These results suggest that photosensitization of U-87MG and Neuro 2a cells by MC 540 involves both type I (free radical mediated) and type II (singlet oxygen mediated) mechanisms. Topics: Animals; Drug Screening Assays, Antitumor; Glioblastoma; Humans; Lipid Peroxidation; Mice; Neuroblastoma; Photosensitizing Agents; Pyrimidinones; Tumor Cells, Cultured | 1997 |
Merocyanine 540 mediated photoirradiation of leukemic cells. In vitro inference on cell survival.
In order to evaluate the selective killing of merocyanine 540 (MC 540) mediated photoirradiation in neoplastic cells, bone narrow cells from children with leukaemia or neuroblastoma and normal children as well as peripheral blood cells and Reh-6 and HL-60 cell lines were studied. Cell suspensions were incubated with MC 540 and exposed to various argon laser 514 nm doses. Cell survival was estimated with trypan blue supravital stain following a 24 h incubation and has been followed in continuous cell cultures of 4 weeks duration. Our results showed that the inhibition of survival of neoplastic haemopoietic cells by laser in the presence of MC 540 is proportional to the MC 540 and photoirradiation doses. A 99.9999% inhibition of Reh-6 and HL-60 was noted at irradiation doses where the corresponding mean survival of normal bone narrow cells was (33.6 +/- 15.5)% and (50.6 +/- 10.7)% respectively. Peripheral blood mononuclear cells were not sensitive to MC 540 mediated photoirradiation. The inhibition of survival of bone marrow metastatic neuroblastoma cells was (69.9 +/- 4.1)%. In conclusion, it seems that MC 540 mediated photoirradiation in neoplastic cells exerts selective cytotoxicity and can be used in ex vivo purging of malignant cells in the bone marrow. Topics: Bone Marrow; Cell Line; Cell Survival; Child; Dose-Response Relationship, Radiation; Hematopoietic Stem Cells; HL-60 Cells; Humans; Kinetics; Leukemia; Light; Neuroblastoma; Photosensitizing Agents; Pyrimidinones; Tumor Cells, Cultured | 1996 |
MC540 induced photosensitization of glioma & neuroblastoma cells.
Binding and photodynamic action of merocyanine 540 (MC540) has been studied in glioma (U-87MG) and neuroblastoma (Neuro 2A) cells as a function of dye concentration, incubation time of cells with MC540 and growth phase of cells. In the plateau phase, U-87MG cells accumulated more MC540 as compared to exponentially growing cells, whereas in Neuro 2A cells the opposite effect was observed. Exponentially growing U-87MG cells were more photosensitive than plateau phase cells. However, the photosensitivity of Neuro 2A cells was not dependent on the growth phase. Thus, MC540 mediated photosensitization may be useful for photodynamic therapy of brain tumours. Topics: Fluorescent Dyes; Glioma; Neuroblastoma; Photochemotherapy; Photosensitizing Agents; Pyrimidinones; Staining and Labeling; Tumor Cells, Cultured | 1994 |
Investigation of the usefulness of the photodynamic reaction for the purging of bone marrow from metastatic neuroblastoma cells.
The neuroblastoma cell lines SK-N-LO, SK-PN-DW and IMR 5 were stained with Merocyanine 540 and exposed to white light. 99% of clonogenic tumour cells were destroyed while 60% of bone marrow progenitor cells survived this treatment. Topics: Bone Marrow; Bone Marrow Purging; Cell Survival; Hematopoietic Stem Cells; Humans; Light; Neuroblastoma; Photochemotherapy; Photosensitizing Agents; Pyrimidinones; Tumor Cells, Cultured | 1993 |
Cytotoxic effects of 6-hydroxydopamine, merocyanine-540 and related compounds on human neuroblastoma and hematopoietic stem cells.
6-Hydroxydopamine(6-OHDA) and Merocyanine-540(MC-540) have been used clinically for purging of neuroblastoma cells prior to autologous bone marrow transplantation. Both substances were found to be more toxic against neuroblastoma cells than against hematopoietic stem cells. The more pronounced cytotoxic effects of 6-OHDA against neuroblastoma cells were not caused by its selective uptake; the rapid autooxidation at physiological pH leads to the formation of H2O2 already in the incubation medium. Cytotoxic effects were not detected in short-time test systems (4 hour chromium-51 release assay) but only after longer incubation periods. In contrast, MC-540 proved to be toxic almost equally in short- and long-time test systems. 4-Hydroxynonenal(4-HNE) that may be formed in the plasma membrane subsequently to photoactivation of MC-540 was only slightly more toxic to neuroblastoma cells than to hematopoietic cells. Although the use of 6-OHDA and MC-540 in bone marrow purging has some limitations, the sensitivity of neuroblastoma cells against reactive oxygen compounds may be exploited more generally for therapy of this tumor. Topics: Aldehydes; Antineoplastic Agents; Cell Survival; Hematopoietic Stem Cells; Humans; Hydrogen Peroxide; Hydroxydopamines; Neuroblastoma; Oxidopamine; Pyrimidinones; Tumor Cells, Cultured | 1989 |
Purging by dye-mediated photosensitization.
Topics: Animals; Bone Marrow; Bone Marrow Transplantation; Hematopoietic Stem Cells; Humans; Leukemia; Leukemia, Experimental; Mice; Neoplastic Stem Cells; Neuroblastoma; Photochemistry; Preoperative Care; Pyrimidinones; Radiation-Sensitizing Agents; Tumor Cells, Cultured; Tumor Stem Cell Assay | 1989 |
Effects of bradykinin, GTP gamma S, R59022 and N-ethylmaleimide on inositol phosphate production in NG108-15 cells.
Accumulation of inositol phosphates in NG108-15 neuroblastoma x glioma hybrid cells, pre-labeled for 24h to equilibrium, was stimulated by bradykinin, guanosine 5'-O-(3-thiotriphosphate) and the diacylglycerol kinase inhibitor R59022. Only the stimulation by bradykinin was inhibited by the bradykinin receptor antagonist [D-Arg0, Hyp3, Phe7, Thi5,8] bradykinin. Neither bradykinin nor R059022 increased the labeling of the inositol phospholipids. The sulfhydryl-alkylating reagent N-ethylmaleimide at 100 microM essentially abolished the stimulation caused by all three agents, possibly by preventing the binding of GTP to a guanine nucleotide-binding regulatory protein of as yet unknown size. Topics: Animals; Bradykinin; Cell Line; Drug Interactions; Ethylmaleimide; Glioma; Guanosine 5'-O-(3-Thiotriphosphate); Guanosine Triphosphate; Hybrid Cells; Inositol; Inositol Phosphates; Kinetics; Mice; Neuroblastoma; Platelet Activating Factor; Pyrimidinones; Rats; Thiazoles; Thionucleotides | 1989 |
Bradykinin-induced translocation of protein kinases C in neuroblastoma NCB-20 cell: dependence on 1,2-diacylglycerol content and free calcium.
Bradykinin (BK)-induced production of 1,2-diacylglycerol (1,2-DG) and translocation of protein kinases C (PKCs) were examined in neuroblastoma-derived hybrid NCB-20 cells. Mass analysis of 1,2-DG exhibited a biphasic increase by 1 microM BK stimulation: the first transient phase and the second broad sustained phase. Among three subspecies of PKC expressed in these cells, types II and III were observed to translocate from cytosol to membrane in response to BK as well as PBt2 by Western blotting analysis. Type II translocated more rapidly and distinctly than type III. However, after treatment with quin 2/AM, the second phase of 1,2-DG formation completely disappeared and PKCs translocation by BK or PBt2 was completely abolished. BK-induced IP3 (1,4,5) formation was temporally consistent with the first transient phase of 1,2-DG formation. These findings suggest that PKCs translocation by BK stimulation is caused by 1,2-DG produced not only via phosphoinositide metabolism, but via other phospholipid breakdown which is Ca2+-dependent. Topics: Animals; Bradykinin; Calcium; Cell Compartmentation; Diglycerides; Glycerides; Humans; Hybrid Cells; Isoenzymes; Neuroblastoma; Phorbol 12,13-Dibutyrate; Protein Kinase C; Pyrimidinones; Thiazoles; Time Factors; Tumor Cells, Cultured | 1989 |
Dye-mediated photosensitization of murine neuroblastoma cells.
The purpose of this study was to determine if photosensitization mediated by the fluorescent dye, merocyanine 540, could be used to preferentially kill murine neuroblastoma cells in simulated autologous remission marrow grafts. Simultaneous exposure of Neuro 2a or NB41A3 neuroblastoma cells to merocyanine 540 and white light reduced the concentration of in vitro-clonogenic tumor cells 50,000-fold. By contrast, the same treatment had little effect on the graft's ability to rescue lethally irradiated syngeneic hosts. Lethally irradiated C57BL/6J X A/J F1 mice transplanted with photosensitized mixtures of neuroblastoma cells and normal marrow cells (1:100 or 1:10) survived without developing neuroblastomas. It is conceivable that merocyanine 540-mediated photosensitization will prove useful for the extracorporeal purging of residual neuroblastoma cells from human autologous remission marrow grafts. Topics: Animals; Bone Marrow; Bone Marrow Transplantation; Cell Line; Cell Survival; Female; Hematopoietic Stem Cells; Mice; Mice, Inbred C57BL; Neuroblastoma; Photochemotherapy; Pyrimidinones; Radiation-Sensitizing Agents; Transplantation, Autologous; Trypsin | 1986 |
Dye-mediated photolysis of human neuroblastoma cells: implications for autologous bone marrow transplantation.
Cells from three different human neuroblastoma cell lines and normal human bone marrow cells were exposed to the lipophilic fluorescent dye, merocyanine 540 (MC 540), and white light. In vitro clonogenic tumor cells were inactivated up to 25,000 times more rapidly than multipotent hematopoietic progenitor cells (CFU-GEMM). It is conceivable that this pronounced difference in sensitivity to MC 540-mediated photolysis can be exploited for the selective killing of residual neuroblastoma cells in autologous remission marrow grafts. Topics: Bone Marrow; Bone Marrow Transplantation; Cell Line; Cell Survival; Hematopoietic Stem Cells; Humans; Neoplastic Stem Cells; Neuroblastoma; Photolysis; Pyrimidinones; Transplantation, Autologous | 1986 |