Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Neoplasia ; 24(2): 109-119, 2022 02.
Article in English | MEDLINE | ID: mdl-34959030

ABSTRACT

BH3 mimetics are promising novel anticancer therapeutics. By selectively inhibiting BCL-2, BCL-xL, or MCL-1 (i.e. ABT-199, A-1331852, S63845) they shift the balance of pro- and anti-apoptotic proteins in favor of apoptosis. As Bromodomain and Extra Terminal (BET) protein inhibitors promote pro-apoptotic rebalancing, we evaluated the potential of the BET inhibitor JQ1 in combination with ABT-199, A-1331852 or S63845 in rhabdomyosarcoma (RMS) cells. The strongest synergistic interaction was identified for JQ1/A-1331852 and JQ1/S63845 co-treatment, which reduced cell viability and long-term clonogenic survival. Mechanistic studies revealed that JQ1 upregulated BIM and NOXA accompanied by downregulation of BCL-xL, promoting pro-apoptotic rebalancing of BCL-2 proteins. JQ1/A-1331852 and JQ1/S63845 co-treatment enhanced this pro-apoptotic rebalancing and triggered BAK- and BAX-dependent apoptosis since a) genetic silencing of BIM, BAK or BAX, b) inhibition of caspase activity with zVAD.fmk and c) overexpression of BCL-2 all rescued JQ1/A-1331852- and JQ1/S63845-induced cell death. Interestingly, NOXA played a different role in both treatments, as genetic silencing of NOXA significantly rescued from JQ1/A-1331852-mediated apoptosis but not from JQ1/S63845-mediated apoptosis. In summary, JQ1/A-1331852 and JQ1/S63845 co-treatment represent new promising therapeutic strategies to synergistically trigger mitochondrial apoptosis in RMS.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Biomimetics , Cell Line, Tumor , Drug Synergism , Humans , Rhabdomyosarcoma
3.
Transl Oncol ; 16: 101313, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34906889

ABSTRACT

The RAS/MEK/ERK genetic axis is commonly altered in rhabdomyosarcoma (RMS), indicating high activity of downstream effector ERK1/2 kinase. Previously, we have demonstrated that inhibition of the RAS/MEK/ERK signaling pathway in RMS is insufficient to induce cell death due to residual pro-survival MCL-1 activity. Here, we show that the combination of ERK1/2 inhibitor Ulixertinib and MCL-1 inhibitor S63845 is highly synergistic and induces apoptotic cell death in RMS in vitro and in vivo. Importantly, Ulixertinib/S63845 co-treatment suppresses long-term survival of RMS cells, induces rapid caspase activation and caspase-dependent apoptosis. Mechanistically, Ulixertinib-mediated upregulation of BIM and BMF in combination with MCL-1 inhibition by S63845 shifts the balance of BCL-2 proteins towards a pro-apoptotic state resulting in apoptosis induction. A genetic silencing approach reveals that BIM, BMF, BAK and BAX are all required for Ulixertinib/S63845-induced apoptosis. Overexpression of BCL-2 rescues cell death triggered by Ulixertinib/S63845 co-treatment, confirming that combined inhibition of ERK1/2 and MCL-1 effectively induces cell death of RMS cells via the intrinsic mitochondrial apoptotic pathway. Thus, this study is the first to demonstrate the cytotoxic potency of co-inhibition of ERK1/2 and MCL-1 for RMS treatment.

4.
Neoplasia ; 23(5): 539-550, 2021 05.
Article in English | MEDLINE | ID: mdl-33971465

ABSTRACT

Burkitt's lymphoma (BL) is a highly aggressive form of B-cell non-Hodgkin's lymphoma. The clinical outcome in children with BL has improved over the last years but the prognosis for adults is still poor, highlighting the need for novel treatment strategies. Here, we report that the combinational treatment with the Smac mimetic BV6 and TRAIL triggers necroptosis in BL when caspases are blocked by zVAD.fmk (TBZ treatment). The sensitivity of BL cells to TBZ correlates with MLKL expression. We demonstrate that necroptotic signaling critically depends on MLKL, since siRNA-induced knockdown and CRISPR/Cas9-mediated knockout of MLKL profoundly protect BL cells from TBZ-induced necroptosis. Conversely, MLKL overexpression in cell lines expressing low levels of MLKL leads to necroptosis induction, which can be rescued by pharmacological inhibitors, highlighting the important role of MLKL for necroptosis execution. Importantly, the methylation status analysis of the MLKL promoter reveals a correlation between methylation and MLKL expression. Thus, MLKL is epigenetically regulated in BL and might serve as a prognostic marker for treatment success of necroptosis-based therapies. These findings have crucial implications for the development of new treatment options for BL.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Mitochondrial Proteins/metabolism , Necroptosis/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis Regulatory Proteins/chemistry , Biological Mimicry , Burkitt Lymphoma/pathology , Cell Death , Cell Line, Tumor , Disease Susceptibility , Gene Expression Regulation, Neoplastic , Humans , Ligands , Mitochondrial Proteins/chemistry , Necroptosis/drug effects , Oligopeptides/metabolism , Oligopeptides/pharmacology , RNA Interference
5.
Cancer Lett ; 490: 76-88, 2020 10 10.
Article in English | MEDLINE | ID: mdl-32679165

ABSTRACT

Rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma, is associated with a low 5-year survival and harsh treatment side effects, underscoring an urgent need for therapy. The unfolded protein response (UPR) is activated in response to endoplasmic reticulum (ER) stress, where three ER stress receptors, IRE1, PERK and ATF6, aim to restore cellular homeostasis. The UPR is pro-tumourigenic in many cancers. In this study, we investigate basal UPR activity in RMS. Basal activation of IRE1 and PERK was observed in RMS cell lines, which was diminished upon addition of the IRE1 RNase inhibitor, MKC8866, or PERK inhibitor, AMGEN44. UPR inhibition caused a reduction in cell viability, cell proliferation and inhibition of long-term colony formation in both subtypes of RMS. Alveolar RMS (ARMS) subtype was highly sensitive to IRE1 inhibition, whereas embryonal RMS (ERMS) subtypes responded more markedly to PERK inhibition. Further investigation revealed a robust activation of senescence upon UPR inhibition. For the first time, the UPR is implicated in RMS biology and phenotype, and inhibition of UPR signalling reduces cell growth, suggesting that the UPR may be a promising target in RMS.


Subject(s)
Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Rhabdomyosarcoma/pathology , Unfolded Protein Response/physiology , eIF-2 Kinase/metabolism , Cell Line, Tumor , Cell Survival , Humans , Rhabdomyosarcoma/metabolism
6.
Oncogene ; 39(19): 3837-3852, 2020 05.
Article in English | MEDLINE | ID: mdl-32161312

ABSTRACT

Remodeling transcription by targeting bromodomain and extraterminal (BET) proteins has emerged as promising anticancer strategy. Here, we identify a novel synergistic interaction of the BET inhibitor JQ1 with the PI3Kα-specific inhibitor BYL719 to trigger mitochondrial apoptosis and to suppress tumor growth in models of rhabdomyosarcoma (RMS). RNA-Seq revealed that JQ1/BYL719 co-treatment shifts the overall balance of BCL-2 family gene expression towards apoptosis and upregulates expression of BMF, BCL2L11 (BIM), and PMAIP1 (NOXA) while downregulating BCL2L1 (BCL-xL). These changes were confirmed by qRT-PCR and western blot analysis. Ingenuity pathway analysis (IPA) of RNA-Seq data followed by validation qRT-PCR and western blot identified MYC and FOXO3a as potential transcription factors (TFs) upstream of the observed gene expression pattern. Immunoprecipitation (IP) studies showed that JQ1/BYL719-stimulated increase in BIM expression enhances the neutralization of antiapoptotic BCL-2, BCL-xL, and MCL-1. This promotes the activation of BAK and BAX and caspase-dependent apoptosis, as (1) individual silencing of BMF, BIM, NOXA, BAK, or BAX, (2) overexpression of BCL-2 or MCL-1 or (3) the caspase inhibitor N-Benzyloxycarbonyl-Val-Ala-Asp(O-Me) fluoromethylketone (zVAD.fmk) all rescue JQ1/BYL719-induced cell death. In conclusion, co-inhibition of BET proteins and PI3Kα cooperatively induces mitochondrial apoptosis by proapoptotic re-balancing of BCL-2 family proteins. This discovery opens exciting perspectives for therapeutic exploitation of BET inhibitors in RMS.


Subject(s)
Mitochondria/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Rhabdomyosarcoma/drug therapy , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/drug effects , Azepines/pharmacology , Bcl-2-Like Protein 11/genetics , Drug Synergism , Gene Expression Regulation, Neoplastic/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Myeloid Cell Leukemia Sequence 1 Protein/genetics , RNA-Seq , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Thiazoles/pharmacology , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , bcl-X Protein/genetics
7.
Int J Cancer ; 146(2): 510-520, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31173656

ABSTRACT

Recent data suggest that rhabdomyosarcoma (RMS) cells might be vulnerable to oxidative stress-induced cell death. Here, we show that RMS are susceptible to cell death induced by Erastin, an inhibitor of the glutamate/cystine antiporter xc- that can increase reactive oxygen species (ROS) production via glutathione (GSH) depletion. Prior to cell death, Erastin caused GSH depletion, ROS production and lipid peroxidation. Importantly, pharmacological inhibitors of lipid peroxidation (i.e., Ferrostatin-1, Liproxstatin-1), ROS scavengers (i.e., α-Tocopherol, GSH) and the iron chelator Deferoxamine inhibited ROS accumulation, lipid peroxidation and cell death, consistent with ferroptosis. Interestingly, the broad-spectrum protein kinase C (PKC) inhibitor Bisindolylmaleimide I as well as the PKCα- and ß-selective inhibitor Gö6976 significantly reduced Erastin-induced cell death. Similarly, genetic knockdown of PKCα significantly protected RMS cells from Erastin-induced cell death. Furthermore, the broad-spectrum nicotinamide adenine dinucleotide phosphate-oxidase (NOX) inhibitor Diphenyleneiodonium and the selective NOX1/4 isoform inhibitor GKT137831 significantly decreased Erastin-stimulated ROS, lipid ROS and cell death. These data provide new insights into the molecular mechanisms of ferroptosis in RMS, contributing to the development of new redox-based treatment strategies.


Subject(s)
Ferroptosis/drug effects , Rhabdomyosarcoma/drug therapy , Apoptosis/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cyclohexylamines/metabolism , Glutathione/metabolism , Humans , Lipid Peroxidation/drug effects , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Phenylenediamines/metabolism , Piperazines/metabolism , Pyrazoles/pharmacology , Pyrazolones , Pyridines/pharmacology , Pyridones , Reactive Oxygen Species/metabolism , Rhabdomyosarcoma/metabolism , alpha-Tocopherol/metabolism
8.
Int J Mol Sci ; 20(3)2019 Jan 27.
Article in English | MEDLINE | ID: mdl-30691192

ABSTRACT

One of the main problems in oncology is the development of drugs that cause the death of cancer cells without damaging normal cells. Another key problem to be solved is to suppress the drug resistance of cancer cells. The third important issue is to provide effective penetration of drug molecules to cancer cells. TRAIL (TNFα-related apoptosis inducing ligand)/Apo2L is a highly selective anticancer agent. However, the recombinant TRAIL protein having high efficiency against cancer cells in vitro was not effective in clinical trials. Recently we have discovered an acquisition of TRAIL resistance by cancer cells in confluent cultures, which is apparently a manifestation of the general phenomenon of multicellular resistance. The aim of this study was to evaluate whether the anticancer effect of the recombinant protein TRAIL in vivo can be improved by the suppression of multicellular TRAIL-resistance using sorafenib and a tumor-penetrating peptide iRGD, c(CRGDKGPDC). The results testified a great increase in the resistance of human fibrosarcoma HT-1080 cells to izTRAIL both in confluent cultures and in spheroids. Sorafenib administered at nontoxic concentration effectively suppressed confluent- or spheroid-mediated TRAIL-resistance of HT-1080 cells in vitro. Sorafenib combined with iRGD significantly improved the anticancer effect of the recombinant protein izTRAIL in HT-1080 human fibrosarcoma grafts in BALB/c nude mice. Consistent with this finding, multicellular TRAIL-resistance may be a reason of inefficacy of izTRAIL alone in vivo. The anticancer effect of the recombinant protein izTRAIL in vivo may be improved in combination with sorafenib, an inhibitor of multicellular TRAIL resistance and iRGD, the tumor-penetrating peptide.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Resistance, Neoplasm/drug effects , Fibrosarcoma/drug therapy , Oligopeptides/administration & dosage , Recombinant Proteins/administration & dosage , Sorafenib/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/pharmacology , Recombinant Proteins/pharmacology , Sorafenib/pharmacology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Xenograft Model Antitumor Assays
9.
Cancer Res ; 78(8): 2000-2013, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29437705

ABSTRACT

Sequencing studies have revealed recurrent mutations in the RAS pathway in rhabdomyosarcoma (RMS). However, RAS effector pathways in RMS are poorly defined. Here, we report that coinhibition of NRAS or MEK plus PI3Kα triggers widespread apoptosis in NRAS-mutated RMS cells. Subtoxic concentrations of the MEK inhibitor MEK162 and the PI3Kα-specific inhibitor BYL719 synergized to trigger apoptosis in NRAS-mutated RMS cells in vitro and in vivoNRAS- or HRAS-mutated cell lines were more vulnerable to MEK162/BYL719 cotreatment than RAS wild-type cell lines, and MEK162/BYL719 cotreatment was more effective to trigger apoptosis in NRAS-mutated than RAS wild-type RMS tumors in vivo We identified BCL-2-modifying factor (BMF) as an inhibitory target of oncogenic NRAS, with either NRAS silencing or MEK inhibition upregulating BMF mRNA and protein levels, which BYL719 further increased. BMF silencing ablated MEK162/BYL719-induced apoptosis. Mechanistic investigations implicated a proapoptotic rebalancing of BCL-2 family members and suppression of cap-dependent translation in apoptotic sensitivity upon MEK162/BYL719 cotreatment. Our results offer a rationale for combining MEK- and PI3Kα-specific inhibitors in clinical treatment of RAS-mutated RMS.Significance: These findings offer a mechanistic rationale for combining MEK- and PI3Kα-specific inhibitors in the clinical treatment of RAS-mutated forms of often untreatable rhabdomyosarcomas. Cancer Res; 78(8); 2000-13. ©2018 AACR.


Subject(s)
Apoptosis , Genes, ras , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mitochondria/metabolism , Mutation , Nuclear Proteins/antagonists & inhibitors , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Transcription Factors/antagonists & inhibitors , Apoptosis/drug effects , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Caspases/metabolism , Cell Line, Tumor , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Gene Silencing , Humans , Protein Biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Caps , Thiazoles/administration & dosage , Thiazoles/pharmacology
10.
Anticancer Drugs ; 28(10): 1118-1125, 2017 11.
Article in English | MEDLINE | ID: mdl-29045271

ABSTRACT

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase that is often overexpressed in rhabdomyosarcoma (RMS). However, its oncogenic and functional role in RMS remains unclear. Therefore, we investigated the antitumor activity of LDK378 (ceritinib), a new second-generation ALK inhibitor approved for patients with ALK-positive non-small-cell lung cancers. Here, we report that LDK378 reduces cell viability and induces cell death in RMS cell lines at low micromolar IC50 concentrations irrespective of ALK expression levels or phosphorylation status. Compared with Karpas 299 non-Hodgkin's lymphoma cells carrying the NPM-ALK fusion gene, RMS cell lines proved to be far less sensitive to LDK378. The broad-range caspase inhibitor zVAD.fmk significantly protects RMS cells from LDK378-mediated cell death, indicating that LDK378 induces caspase-dependent apoptotic cell death. Before the onset of apoptosis, LDK378 reduces phosphorylation of AKT, S6 ribosomal protein, STAT3 and - to a lesser extent - phosphorylation of ERK, showing that it suppresses key survival pathways. Importantly, we identify a synergistic induction of cell death by combining subtoxic concentrations of LDK378 with the multitargeting kinase inhibitor sorafenib. Calculation of the combination index confirmed that this interaction is synergistic. Also, LDK378 cooperates with sorafenib to significantly reduce colony formation of RMS cells, showing that this combination affects long-term clonogenic growth. In conclusion, LDK378 induces caspase-dependent apoptotic cell death in RMS cells independent of their ALK status and synergizes at subtoxic concentrations with sorafenib to induce cell death. These findings have important implications for the use of LDK378 in RMS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/enzymology , Sulfones/pharmacology , Anaplastic Lymphoma Kinase , Animals , Cell Death/drug effects , Cell Line, Tumor , Drug Synergism , Humans , Niacinamide/administration & dosage , Niacinamide/pharmacology , Phenylurea Compounds/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Rhabdomyosarcoma/pathology , Sorafenib , Sulfones/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...