Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(8)2022 Apr 18.
Article in English | MEDLINE | ID: mdl-35454943

ABSTRACT

The MST1R/RON receptor tyrosine kinase is a homologue of the more well-known MET receptor. Like MET, RON orchestrates cell signaling pathways that promote oncogenesis and enable cancer cell survival; however, it has a more unique role in the regulation of inflammation. RON was originally described as a transmembrane receptor expressed on tissue resident macrophages and various epithelial cells. RON is overexpressed in a variety of cancers and its activation modifies multiple signaling pathways with resultant changes in epithelial and immune cells which together modulate oncogenic phenotypes. While several RON isoforms have been identified with differences in structure, activation, and pathway regulation, increased RON expression and/or activation is consistently associated with worse outcomes. Tyrosine kinase inhibitors targeting RON have been developed, making RON an actionable therapeutic target.

2.
Clin Cancer Res ; 27(7): 2100-2110, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33451980

ABSTRACT

PURPOSE: Pancreatic cancer is an aggressive disease associated with a poor 5-year overall survival. Most patients are ineligible for surgery due to late diagnosis and are treated primarily with chemotherapy with very limited success. Pancreatic cancer is relatively insensitive to chemotherapy due to multiple factors, including reduced bioavailability of drugs to tumor cells. One strategy to improve drug efficacy with reduced toxicity is the development of antibody-drug conjugates (ADC), which have now been used successfully to treat both solid and liquid tumors. Here, we evaluate the efficacy of TR1801-ADC, a newly developed ADC composed of a MET antibody conjugated to the highly potent pyrrolobenzodiazepine toxin-linker, tesirine. EXPERIMENTAL DESIGN: We first evaluated MET expression and subcellular localization in pancreatic cancer cell lines, human tumors, and patient-derived xenografts (PDX). We then tested TR1801-ADC efficacy in vitro in pancreatic cancer cell lines. Preclinical evaluation of TR1801-ADC efficacy was conducted on PDXs selected on the basis of their MET expression level. RESULTS: We show that MET is highly expressed and located at the plasma membrane of pancreatic cancer cells. We found that TR1801-ADC induces a specific cytotoxicity in pancreatic cancer cell lines and a profound tumor growth inhibition, even in a gemcitabine-resistant tumor. We also noted synergism between TR1801-ADC and gemcitabine in vitro and an improved response to the combination in vivo. CONCLUSIONS: Together, these results suggest the promise of agents such as TR1801-ADC as a novel approach to the treatment of pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Immunoconjugates/therapeutic use , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/immunology , Animals , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm , Humans , Male , Mice , Pancreatic Neoplasms/mortality , Proto-Oncogene Proteins c-met/analysis , Xenograft Model Antitumor Assays , Gemcitabine
3.
Oncogene ; 38(28): 5599-5611, 2019 07.
Article in English | MEDLINE | ID: mdl-30967626

ABSTRACT

The MST1R (RON) kinase is overexpressed in >80% of human pancreatic cancers, but its role in pancreatic carcinogenesis is unknown. In this study, we examined the relevance of Mst1r kinase to Kras driven pancreatic carcinogenesis using genetically engineered mouse models. In the setting of mutant Kras, Mst1r overexpression increased acinar-ductal metaplasia (ADM), accelerated the progression of pancreatic intraepithelial neoplasia (PanIN), and resulted in the accumulation of (mannose receptor C type 1) MRC1+, (arginase 1) Arg+ macrophages in the tumor microenvironment. Conversely, absence of a functional Mst1r kinase slowed PanIN initiation, resulted in smaller tumors, prolonged survival and a reduced tumor-associated macrophage content. Mst1r expression was associated with increased production of its ligand Mst1, and in orthotopic models, suppression of Mst1 expression resulted in reduced tumor size, changes in macrophage polarization and enhanced T cell infiltration. This study demonstrates the functional significance of Mst1r during pancreatic cancer initiation and progression. Further, it provides proof of concept that targeting Mst1r can modulate pancreatic cancer growth and the microenvironment. This study provides further rationale for targeting Mst1r as a therapeutic strategy.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Epithelial Cells/pathology , Macrophages/pathology , Pancreatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Carcinoma, Pancreatic Ductal/enzymology , Disease Progression , Female , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/enzymology , Proof of Concept Study , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Tumor Microenvironment
4.
Anticancer Res ; 36(9): 4459-66, 2016 09.
Article in English | MEDLINE | ID: mdl-27630282

ABSTRACT

BACKGROUND/AIM: Somatostatine receptors subtype 2 (SSTR2) are regarded as a potential target in neuroblastoma (NB) for imaging and promising therapeutic approaches. The purpose of this study was to evaluate and compare the SSTR2 status by (68)Ga-[tetraxetan-D-Phe1, Tyr3]-octreotide ((68)Ga-DOTATOC) positron-emission tomography (PET) and the tumour metabolic activity by (18)F-fluorodeoxyglucose (FDG) PET in different experimental models of NB. MATERIALS AND METHODS: Three cell lines of human NB with different levels of expression of SSTR2 were grafted into nude mice. Animals were imaged with FDG and (68)Ga-DOTATOC and the maximum standardized uptake value (SUVmax) was determined to quantify tracer uptake. Ex vivo biodistribution of (68)Ga-DOTATOC and immunohistochemical analysis of NB xenografts were performed. RESULTS: Compared with FDG, the SUVmax of (68)Ga-DOTATOC uptake by the tumour was lower but the ratio to background was higher; there was a strong positive correlation between SUVmax values observed with the two tracers (r(2)=0.65). Sorting the cell lines according to uptake of FDG or (68)Ga-DOTATOC, injected activity per gram of tissue, Ki67 index or expression of SSTR2 assessed visually led to the same classification. CONCLUSION: (68)Ga-DOTATOC allows preclinical imaging of NB according to the intensity of the expression of SSTR2. In contrast with what has been reported for neuroendocrine tumours, in this NB model, the (68)Ga-DOTATOC uptake was positively correlated with FDG uptake and with Ki67 index, usual markers of tumour aggressiveness. If confirmed in humans, this result would favour a theranostic application of (68)Ga-DOTATOC in NB, even in advanced stages.


Subject(s)
Brain Neoplasms/diagnostic imaging , Fluorodeoxyglucose F18/chemistry , Neuroblastoma/diagnostic imaging , Octreotide/analogs & derivatives , Organometallic Compounds/chemistry , Positron-Emission Tomography , Animals , Cell Line, Tumor , Disease Models, Animal , Gallium Radioisotopes/chemistry , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Octreotide/chemistry , Receptors, Somatostatin/metabolism , Tissue Distribution
6.
Clin Cancer Res ; 21(14): 3327-39, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25805801

ABSTRACT

PURPOSE: Activating ALK mutations are present in almost 10% of primary neuroblastomas and mark patients for treatment with small-molecule ALK inhibitors in clinical trials. However, recent studies have shown that multiple mechanisms drive resistance to these molecular therapies. We anticipated that detailed mapping of the oncogenic ALK-driven signaling in neuroblastoma can aid to identify potential fragile nodes as additional targets for combination therapies. EXPERIMENTAL DESIGN: To achieve this goal, transcriptome profiling was performed in neuroblastoma cell lines with the ALK(F1174L) or ALK(R1275Q) hotspot mutations, ALK amplification, or wild-type ALK following pharmacologic inhibition of ALK using four different compounds. Next, we performed cross-species genomic analyses to identify commonly transcriptionally perturbed genes in MYCN/ALK(F1174L) double transgenic versus MYCN transgenic mouse tumors as compared with the mutant ALK-driven transcriptome in human neuroblastomas. RESULTS: A 77-gene ALK signature was established and successfully validated in primary neuroblastoma samples, in a neuroblastoma cell line with ALK(F1174L) and ALK(R1275Q) regulable overexpression constructs and in other ALKomas. In addition to the previously established PI3K/AKT/mTOR, MAPK/ERK, and MYC/MYCN signaling branches, we identified that mutant ALK drives a strong upregulation of MAPK negative feedback regulators and upregulates RET and RET-driven sympathetic neuronal markers of the cholinergic lineage. CONCLUSIONS: We provide important novel insights into the transcriptional consequences and the complexity of mutant ALK signaling in this aggressive pediatric tumor. The negative feedback loop of MAPK pathway inhibitors may affect novel ALK inhibition therapies, whereas mutant ALK induced RET signaling can offer novel opportunities for testing ALK-RET oriented molecular combination therapies.


Subject(s)
Alkaline Phosphatase/genetics , Drug Resistance, Neoplasm/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Targeted Therapy/methods , Neuroblastoma/genetics , Proto-Oncogene Proteins c-ret/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Feedback, Physiological , Humans , Mice , Mice, Transgenic , Neuroblastoma/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Transcriptome , Up-Regulation
7.
Oncotarget ; 6(1): 26-42, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25622104

ABSTRACT

The gene encoding the LIM and SH3 domain protein (LASP1) was cloned two decades ago from a cDNA library of breast cancer metastases. As the first protein of a class comprising one N-terminal LIM and one C-terminal SH3 domain, LASP1 founded a new LIM-protein subfamily of the nebulin group. Since its discovery LASP1 proved to be an extremely versatile protein because of its exceptional structure allowing interaction with various binding partners, its ubiquitous expression in normal tissues, albeit with distinct expression patterns, and its ability to transmit signals from the cytoplasm into the nucleus. As a result, LASP1 plays key roles in cell structure, physiological processes, and cell signaling. Furthermore, LASP1 overexpression contributes to cancer aggressiveness hinting to a potential value of LASP1 as a cancer biomarker. In this review we summarize published data on structure, regulation, function, and expression pattern of LASP1, with a focus on its role in human cancer and as a biomarker protein. In addition, we provide a comprehensive transcriptome analysis of published microarrays (n=2,780) that illustrates the expression profile of LASP1 in normal tissues and its overexpression in a broad range of human cancer entities.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/metabolism , Cytoskeletal Proteins/metabolism , LIM Domain Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Animals , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Cytoplasm/metabolism , Cytoskeleton/metabolism , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Mice , Parietal Cells, Gastric/cytology , Protein Structure, Tertiary , Signal Transduction , Species Specificity
8.
Oncotarget ; 5(9): 2688-702, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24811913

ABSTRACT

Activating mutations of the ALK (Anaplastic lymphoma Kinase) gene have been identified in sporadic and familial cases of neuroblastoma, a cancer of early childhood arising from the sympathetic nervous system (SNS). To decipher ALK function in neuroblastoma predisposition and oncogenesis, we have characterized knock-in (KI) mice bearing the two most frequent mutations observed in neuroblastoma patients. A dramatic enlargement of sympathetic ganglia is observed in AlkF1178L mice from embryonic to adult stages associated with an increased proliferation of sympathetic neuroblasts from E14.5 to birth. In a MYCN transgenic context, the F1178L mutation displays a higher oncogenic potential than the R1279Q mutation as evident from a shorter latency of tumor onset. We show that tumors expressing the R1279Q mutation are sensitive to ALK inhibition upon crizotinib treatment. Furthermore, our data provide evidence that activated ALK triggers RET upregulation in mouse sympathetic ganglia at birth as well as in murine and human neuroblastoma. Using vandetanib, we show that RET inhibition strongly impairs tumor growth in vivo in both MYCN/KI AlkR1279Q and MYCN/KI AlkF1178L mice. Altogether, our findings demonstrate the critical role of activated ALK in SNS development and pathogenesis and identify RET as a therapeutic target in ALK mutated neuroblastoma.


Subject(s)
Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Mutation/genetics , Neuroblastoma/genetics , Neurogenesis , Proto-Oncogene Proteins c-ret/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Amino Acid Sequence , Anaplastic Lymphoma Kinase , Animals , Base Sequence , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Southern , Blotting, Western , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Integrases , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , N-Myc Proto-Oncogene Protein , Neuroblastoma/metabolism , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-ret/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transcriptional Activation , Tumor Cells, Cultured
9.
PLoS One ; 8(8): e72182, 2013.
Article in English | MEDLINE | ID: mdl-23991058

ABSTRACT

Neuroblastoma is a pediatric cancer of the peripheral nervous system in which structural chromosome aberrations are emblematic of aggressive tumors. In this study, we performed an in-depth analysis of somatic rearrangements in two neuroblastoma cell lines and two primary tumors using paired-end sequencing of mate-pair libraries and RNA-seq. The cell lines presented with typical genetic alterations of neuroblastoma and the two tumors belong to the group of neuroblastoma exhibiting a profile of chromothripsis. Inter and intra-chromosomal rearrangements were identified in the four samples, allowing in particular characterization of unbalanced translocations at high resolution. Using complementary experiments, we further characterized 51 rearrangements at the base pair resolution that revealed 59 DNA junctions. In a subset of cases, complex rearrangements were observed with templated insertion of fragments of nearby sequences. Although we did not identify known particular motifs in the local environment of the breakpoints, we documented frequent microhomologies at the junctions in both chromothripsis and non-chromothripsis associated breakpoints. RNA-seq experiments confirmed expression of several predicted chimeric genes and genes with disrupted exon structure including ALK, NBAS, FHIT, PTPRD and ODZ4. Our study therefore indicates that both non-homologous end joining-mediated repair and replicative processes may account for genomic rearrangements in neuroblastoma. RNA-seq analysis allows the identification of the subset of abnormal transcripts expressed from genomic rearrangements that may be involved in neuroblastoma oncogenesis.


Subject(s)
Chromosome Aberrations , Chromosome Breakpoints , Gene Rearrangement/genetics , Neuroblastoma/genetics , Translocation, Genetic/genetics , Acid Anhydride Hydrolases/genetics , Anaplastic Lymphoma Kinase , Base Sequence , Cell Line, Tumor , Child, Preschool , DNA Copy Number Variations , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Neuroblastoma/pathology , Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Transcription, Genetic
10.
Cancer Res ; 73(1): 195-204, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23139213

ABSTRACT

Activating mutations of the ALK gene have been identified in sporadic and familial cases of neuroblastoma (NB), a cancer of the peripheral nervous system, and are thought to be the primary mechanism of oncogenic activation of this receptor in this pediatric neoplasm. To address the possibility that ALK activation may occur through genomic rearrangements as detected in other cancers, we first took advantage of high-resolution array-comparative genomic hybridization to search for ALK rearrangements in NB samples. Using complementary experiments by capture/paired-end sequencing and FISH experiments, various types of rearrangements were fully characterized, including partial gains or amplifications, in several NB cell lines and primary tumors. In the CLB-Bar cell line, we described a genomic rearrangement associated with an amplification of the ALK locus, leading to the expression of a 170 kDa protein lacking part of the extracellular domain encoded by exons 4 to 11, named ALK(Δ4-11). Analysis of genomic DNA from the tumor at diagnosis and relapse revealed that the ALK gene was amplified at diagnosis but that the rearranged ALK allele was observed at the relapse stage only, suggesting that it may be implicated in tumor aggressiveness. Consistently, oncogenic and tumorigenic properties of the ALK(Δ4-11) variant were shown after stable expression in NIH3T3 cells. Moreover, we documented an increased constitutive kinase activity of this variant, as well as an impaired maturation and retention into intracellular compartments. These results indicate that genomic rearrangements constitute an alternative mechanism to ALK point mutations resulting in receptor activation.


Subject(s)
Gene Rearrangement/genetics , Neuroblastoma/genetics , Receptor Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Cell Line, Tumor , Comparative Genomic Hybridization , Humans , Immunoblotting , Immunoprecipitation , In Situ Hybridization, Fluorescence
11.
PLoS One ; 7(3): e33581, 2012.
Article in English | MEDLINE | ID: mdl-22479414

ABSTRACT

Recently, activating mutations of the full length ALK receptor, with two hot spots at positions F1174 and R1275, have been characterized in sporadic cases of neuroblastoma. Here, we report similar basal patterns of ALK phosphorylation between the neuroblastoma IMR-32 cell line, which expresses only the wild-type receptor (ALK(WT)), and the SH-SY5Y cell line, which exhibits a heterozygous ALK F1174L mutation and expresses both ALK(WT) and ALK(F1174L) receptors. We demonstrate that this lack of detectable increased phosphorylation in SH-SY5Y cells is a result of intracellular retention and proteasomal degradation of the mutated receptor. As a consequence, in SH-SY5Y cells, plasma membrane appears strongly enriched for ALK(WT) whereas both ALK(WT) and ALK(F1174L) were present in intracellular compartments. We further explored ALK receptor trafficking by investigating the effect of agonist and antagonist mAb (monoclonal antibodies) on ALK internalization and down-regulation, either in SH-SY5Y cells or in cells expressing only ALK(WT). We observe that treatment with agonist mAbs resulted in ALK internalization and lysosomal targeting for receptor degradation. In contrast, antagonist mAb induced ALK internalization and recycling to the plasma membrane. Importantly, we correlate this differential trafficking of ALK in response to mAb with the recruitment of the ubiquitin ligase Cbl and ALK ubiquitylation only after agonist stimulation. This study provides novel insights into the mechanisms regulating ALK trafficking and degradation, showing that various ALK receptor pools are regulated by proteasome or lysosome pathways according to their intracellular localization.


Subject(s)
Antibodies, Monoclonal/pharmacology , Down-Regulation , Neuroblastoma/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase , Base Sequence , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/genetics , Enzyme Activation/drug effects , Humans , Mutation , Neuroblastoma/genetics , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Transport/drug effects , Receptor Protein-Tyrosine Kinases/agonists , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...