Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
FASEB J ; 34(8): 11101-11114, 2020 08.
Article in English | MEDLINE | ID: mdl-32623799

ABSTRACT

The majority of anti-cancer therapies target the proliferating tumor cells, while the tumor stroma, principally unaffected, survives, and provide a niche for surviving tumor cells. Combining tumor cell and stroma-targeting therapies thus have a potential to improve patient outcome. The neuroblastoma stroma contains cancer-associated fibroblasts expressing microsomal prostaglandin E synthase-1 (mPGES-1). mPGES-1-derived prostaglandin E2 (PGE2 ) is known to promote tumor growth through increased proliferation and survival of tumor cells, immune suppression, angiogenesis, and therapy resistance, and we, therefore, hypothesize that mPGES-1 constitutes an interesting stromal target. Here, we aimed to develop a relevant in vitro model to study combination therapies. Co-culturing of neuroblastoma and fibroblast cells in 3D tumor spheroids mimic neuroblastoma tumors with regard to the cyclooxygenase/mPGES-1/PGE2 pathway. Using the spheroid model, we show that the inhibition of fibroblast-derived mPGES-1 enhanced the cytotoxic effect of doxorubicin and vincristine and significantly reduced tumor cell viability and spheroid growth. Cyclic treatment with vincristine in combination with an mPGES-1 inhibitor abrogated cell repopulation. Moreover, inhibition of mPGES-1 potentiated the cytotoxic effect of vincristine on established neuroblastoma allografts in mice. In conclusion, we established a 3D neuroblastoma model, highlighting the potential of combining stromal targeting of mPGES-1 with tumor cell targeting drugs like vincristine.


Subject(s)
Antineoplastic Agents/pharmacology , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Drug Delivery Systems/methods , Drug Evaluation, Preclinical/methods , Female , Humans , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neuroblastoma/metabolism , Prostaglandin-E Synthases/metabolism
2.
Adv Exp Med Biol ; 1161: 89-100, 2019.
Article in English | MEDLINE | ID: mdl-31562624

ABSTRACT

The importance of prostaglandin E2 in cancer progression is well established, but research on its role in cancer has so far mostly been focused on epithelial cancer in adults while the knowledge about the contribution of prostaglandin E2 to childhood malignancies is limited. Neuroblastoma, an extracranial solid tumor of the sympathetic nervous system, mainly affects young children. Patients with tumors classified as high-risk have poor survival despite receiving intensive treatment, illustrating a need for new treatments complimenting existing ones. The basis of neuroblastoma treatment e.g. chemotherapy and radiation therapy, target the proliferating genetically unstable tumor cells leading to treatment resistance and relapses. The tumor microenvironment is an avenue, still to a great extent, unexplored and lacking effective targeted therapies. Cancer-associated fibroblasts is the main source of prostaglandin E2 in neuroblastoma contributing to angiogenesis, immunosuppression and tumor growth. Prostaglandin E2 is formed from its precursor arachidonic acid in a two-step enzymatic reaction. Arachidonic acid is first converted by cyclooxygenases into prostaglandin H2 and then further converted by microsomal prostaglandin E synthase-1 into prostaglandin E2. We believe targeting of microsomal prostaglandin E synthase-1 in cancer-associated fibroblasts will be an effective future therapeutic strategy in fighting neuroblastoma.


Subject(s)
Dinoprostone , Neuroblastoma , Prostaglandin-E Synthases , Prostaglandin-Endoperoxide Synthases , Antineoplastic Agents/therapeutic use , Arachidonic Acid/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Neuroblastoma/physiopathology , Prostaglandin-E Synthases/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Tumor Microenvironment
3.
EBioMedicine ; 32: 84-92, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29804818

ABSTRACT

Despite recent progress in diagnosis and treatment, survival for children with high-risk metastatic neuroblastoma is still poor. Prostaglandin E2 (PGE2)-driven inflammation promotes tumor growth, immune suppression, angiogenesis and resistance to established cancer therapies. In neuroblastoma, cancer-associated fibroblasts (CAFs) residing in the tumor microenvironment are the primary source of PGE2. However, clinical targeting of PGE2 with current non-steroidal anti-inflammatory drugs or cyclooxygenase inhibitors has been limited due to risk of adverse side effects. By specifically targeting microsomal prostaglandin E synthase-1 (mPGES-1) activity with a small molecule inhibitor we could block CAF-derived PGE2 production leading to reduced tumor growth, impaired angiogenesis, inhibited CAF migration and infiltration, reduced tumor cell proliferation and a favorable shift in the M1/M2 macrophage ratio. In this study, we provide proof-of-principle of the benefits of targeting mPGES-1 in neuroblastoma, applicable to a wide variety of tumors. This non-toxic single drug treatment targeting infiltrating stromal cells opens up for combination treatment options with established cancer therapies.


Subject(s)
Inflammation/drug therapy , Neovascularization, Pathologic/drug therapy , Neuroblastoma/drug therapy , Prostaglandin-E Synthases/genetics , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclooxygenase Inhibitors/administration & dosage , Dinoprostone/genetics , Dinoprostone/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Inflammation/genetics , Inflammation/pathology , Microsomes/drug effects , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neuroblastoma/genetics , Neuroblastoma/pathology , Prostaglandin-E Synthases/antagonists & inhibitors , Tumor Microenvironment/drug effects
4.
Genes Dev ; 31(10): 1036-1053, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28637693

ABSTRACT

We recently identified pathogenic KIF1Bß mutations in sympathetic nervous system malignancies that are defective in developmental apoptosis. Here we deleted KIF1Bß in the mouse sympathetic nervous system and observed impaired sympathetic nervous function and misexpression of genes required for sympathoadrenal lineage differentiation. We discovered that KIF1Bß is required for nerve growth factor (NGF)-dependent neuronal differentiation through anterograde transport of the NGF receptor TRKA. Moreover, pathogenic KIF1Bß mutations identified in neuroblastoma impair TRKA transport. Expression of neuronal differentiation markers is ablated in both KIF1Bß-deficient mouse neuroblasts and human neuroblastomas that lack KIF1Bß. Transcriptomic analyses show that unfavorable neuroblastomas resemble mouse sympathetic neuroblasts lacking KIF1Bß independent of MYCN amplification and the loss of genes neighboring KIF1B on chromosome 1p36. Thus, defective precursor cell differentiation, a common trait of aggressive childhood malignancies, is a pathogenic effect of KIF1Bß loss in neuroblastomas. Furthermore, neuropathy-associated KIF1Bß mutations impede cargo transport, providing a direct link between neuroblastomas and neurodegeneration.


Subject(s)
Cell Differentiation/genetics , Kinesins/genetics , Kinesins/metabolism , Neuroblastoma/genetics , Neurons/cytology , Receptor, trkA/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Regulation, Developmental , Gene Silencing , Mutation , Neuroblastoma/physiopathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , PC12 Cells , Rats , Signal Transduction/genetics , Sympathetic Nervous System/cytology , ras Proteins/genetics
5.
Cell Rep ; 16(4): 979-993, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27396325

ABSTRACT

MYCN amplification and MYC signaling are associated with high-risk neuroblastoma with poor prognosis. Treating these tumors remains challenging, although therapeutic approaches stimulating differentiation have generated considerable interest. We have previously shown that the MYCN-regulated miR-17∼92 cluster inhibits neuroblastoma differentiation by repressing estrogen receptor alpha. Here, we demonstrate that this microRNA (miRNA) cluster selectively targets several members of the nuclear hormone receptor (NHR) superfamily, and we present a unique NHR signature associated with the survival of neuroblastoma patients. We found that suppressing glucocorticoid receptor (GR) expression in MYCN-driven patient and mouse tumors was associated with an undifferentiated phenotype and decreased survival. Importantly, MYCN inhibition and subsequent reactivation of GR signaling promotes neural differentiation and reduces tumor burden. Our findings reveal a key role for the miR-17∼92-regulated NHRs in neuroblastoma biology, thereby providing a potential differentiation approach for treating neuroblastoma patients.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/genetics , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Nude , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Receptors, Glucocorticoid/genetics , Signal Transduction/genetics
6.
Proc Natl Acad Sci U S A ; 112(26): 8070-5, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26080408

ABSTRACT

The majority of solid tumors are presented with an inflammatory microenvironment. Proinflammatory lipid mediators including prostaglandin E2 (PGE2) contribute to the establishment of inflammation and have been linked to tumor growth and aggressiveness. Here we show that high-risk neuroblastoma with deletion of chromosome 11q represents an inflammatory subset of neuroblastomas. Analysis of enzymes involved in the production of proinflammatory lipid mediators showed that 11q-deleted neuroblastoma tumors express high levels of microsomal prostaglandin E synthase-1 (mPGES-1) and elevated levels of PGE2. High mPGES-1 expression also corresponded to poor survival of neuroblastoma patients. Investigation of the tumor microenvironment showed high infiltration of tumor-promoting macrophages with high expression of the M2-polarization markers CD163 and CD206. mPGES-1-expressing cells in tumors from different subtypes of neuroblastoma showed differential expression of one or several cancer-associated fibroblast markers such as vimentin, fibroblast activation protein α, α smooth muscle actin, and PDGF receptor ß. Importantly, inhibition of PGE2 production with diclofenac, a nonselective COX inhibitor, resulted in reduced tumor growth in an in vivo model of 11q-deleted neuroblastoma. Collectively, these results suggest that PGE2 is involved in the tumor microenvironment of specific neuroblastoma subgroups and indicate that therapeutic strategies using existing anti-inflammatory drugs in combination with current treatment should be considered for certain neuroblastomas.


Subject(s)
Dinoprostone/metabolism , Inflammation/metabolism , Intramolecular Oxidoreductases/metabolism , Neuroblastoma/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Chromosome Deletion , Chromosomes, Human, Pair 11 , Disease Models, Animal , Humans , Inflammation/enzymology , Inflammation/pathology , Intramolecular Oxidoreductases/genetics , Mice , Neuroblastoma/enzymology , Neuroblastoma/pathology , Prostaglandin-E Synthases , RNA, Messenger/genetics , Tumor Microenvironment
7.
PLoS One ; 7(1): e29331, 2012.
Article in English | MEDLINE | ID: mdl-22276108

ABSTRACT

BACKGROUND: Prostaglandin E(2) (PGE(2)) is an important mediator in tumor-promoting inflammation. High expression of cyclooxygenase-2 (COX-2) has been detected in the embryonic childhood tumor neuroblastoma, and treatment with COX inhibitors significantly reduces tumor growth. Here, we have investigated the significance of a high COX-2 expression in neuroblastoma by analysis of PGE(2) production, the expression pattern and localization of PGE(2) receptors and intracellular signal transduction pathways activated by PGE(2). PRINCIPAL FINDINGS: A high expression of the PGE(2) receptors, EP1, EP2, EP3 and EP4 in primary neuroblastomas, independent of biological and clinical characteristics, was detected using immunohistochemistry. In addition, mRNA and protein corresponding to each of the receptors were detected in neuroblastoma cell lines. Immunofluorescent staining revealed localization of the receptors to the cellular membrane, in the cytoplasm, and in the nuclear compartment. Neuroblastoma cells produced PGE(2) and stimulation of serum-starved neuroblastoma cells with PGE(2) increased the intracellular concentration of calcium and cyclic AMP with subsequent phosphorylation of Akt. Addition of 16,16-dimethyl PGE(2) (dmPGE(2)) increased cell viability in a time, dose- and cell line-dependent manner. Treatment of neuroblastoma cells with a COX-2 inhibitor resulted in a diminished cell growth and viability that was reversed by the addition of dmPGE(2). Similarly, PGE(2) receptor antagonists caused a decrease in neuroblastoma cell viability in a dose-dependent manner. CONCLUSIONS: These findings demonstrate that PGE(2) acts as an autocrine and/or paracrine survival factor for neuroblastoma cells. Hence, specific targeting of PGE(2) signaling provides a novel strategy for the treatment of childhood neuroblastoma through the inhibition of important mediators of tumor-promoting inflammation.


Subject(s)
16,16-Dimethylprostaglandin E2/pharmacology , Autocrine Communication/drug effects , Dinoprostone/metabolism , Neuroblastoma/metabolism , Biphenyl Compounds/pharmacology , Blotting, Western , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Immunohistochemistry , In Vitro Techniques , Phosphorylation/drug effects , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry , Thiophenes/pharmacology , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL