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










Publication year range
1.
bioRxiv ; 2024 May 26.
Article in English | MEDLINE | ID: mdl-38826212

ABSTRACT

A blood test that enables surveillance for early-stage pancreatic ductal adenocarcinoma (PDAC) is an urgent need. Independent laboratories have reported PDAC biomarkers that could improve biomarker performance over CA19-9 alone, but the performance of the previously reported biomarkers in combination is not known. Therefore, we conducted a coordinated case/control study across multiple laboratories using common sets of blinded training and validation samples (132 and 295 plasma samples, respectively) from PDAC patients and non-PDAC control subjects representing conditions under which surveillance occurs. We analyzed the training set to identify candidate biomarker combination panels using biomarkers across laboratories, and we applied the fixed panels to the validation set. The panels identified in the training set, CA19-9 with CA199.STRA, LRG1, TIMP-1, TGM2, THSP2, ANG, and MUC16.STRA, achieved consistent performance in the validation set. The panel of CA19-9 with the glycan biomarker CA199.STRA improved sensitivity from 0.44 with 0.98 specificity for CA19-9 alone to 0.71 with 0.98 specificity (p < 0.001, 1000-fold bootstrap). Similarly, CA19-9 combined with the protein biomarker LRG1 and CA199.STRA improved specificity from 0.16 with 0.94 sensitivity for CA19-9 to 0.65 with 0.89 sensitivity (p < 0.001, 1000-fold bootstrap). We further validated significantly improved performance using biomarker panels that did not include CA19-9. This study establishes the effectiveness of a coordinated study of previously discovered biomarkers and identified panels of those biomarkers that significantly increased the sensitivity and specificity of early-stage PDAC detection in a rigorous validation trial.

2.
Front Psychol ; 12: 720427, 2021.
Article in English | MEDLINE | ID: mdl-34867599

ABSTRACT

Describing sets in terms of a two-valued variable, either value can be chosen: exam results may be referred to by pass rates or fail rates. What determines such framing choices? Building on work by McKenzie and colleagues on reference points in the production and interpretation of framed information, we investigate two determinants of frame choice. One is that speakers tend to focus on the component that has increased vis-à-vis a previous state, the other is the tendency to choose the component larger than 50%. We propose to view reference points as pointing to different kinds of communicative relevance. Hence the use of the previous state and the 50% reference points by speakers is not just a function of the information, but is co-determined by a communicative cue in the context: the question being asked about this information. This line of thought is supported by two experiments containing items offering two-sided distribution information at two points in time. Our first experiment employs a static task, requiring a description of the most recent situation. The second experiment uses a dynamic task, asking participants to describe the development between the two time points. We hypothesize that in static tasks the component size is the strongest frame choice determinant, while in dynamic tasks frame choice is mainly driven by whether a component has increased. The experiments consist of 16 different scenarios, both with symmetrical contrasts (i.e., dogs vs. cats) and with asymmetrical ones (i.e., winning vs. losing). Both experiments support the hypotheses. In the static task, the size effect is the only consistent effect; in the dynamic task, the effect of direction of change is much larger than that of size. This pattern of differences between size and change effects applies across symmetrical and asymmetrical contrasts. Our experiments shed light on cognitive and communicative regularities involved in the production of framed messages: people do tend to prefer larger and increasing components when choosing a frame, but the relative strength of both these preferences depends on the communicative task.

3.
Clin Cancer Res ; 27(1): 226-236, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33093149

ABSTRACT

PURPOSE: A subset of pancreatic ductal adenocarcinomas (PDACs) is highly resistant to systemic chemotherapy, but no markers are available in clinical settings to identify this subset. We hypothesized that a glycan biomarker for PDACs called sialylated tumor-related antigen (sTRA) could be used for this purpose. EXPERIMENTAL DESIGN: We tested for differences between PDACs classified by glycan expression in multiple systems: sets of cell lines, organoids, and isogenic cell lines; primary tumors; and blood plasma from human subjects. RESULTS: The sTRA-expressing models tended to have stem-like gene expression and the capacity for mesenchymal differentiation, in contrast to the nonexpressing models. The sTRA cell lines also had significantly increased resistance to seven different chemotherapeutics commonly used against pancreatic cancer. Patients with primary tumors that were positive for a gene expression classifier for sTRA received no statistically significant benefit from adjuvant chemotherapy, in contrast to those negative for the signature. In another cohort, based on direct measurements of sTRA in tissue microarrays, the patients who were high in sTRA again had no statistically significant benefit from adjuvant chemotherapy. Furthermore, a blood plasma test for the sTRA glycan identified the PDACs that showed rapid relapse following neoadjuvant chemotherapy. CONCLUSIONS: This research demonstrates that a glycan biomarker could have value to detect chemotherapy-resistant PDAC in clinical settings. This capability could aid in the development of stratified treatment plans and facilitate biomarker-guided trials targeting resistant PDAC.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/blood , Carcinoma, Pancreatic Ductal/drug therapy , Neoplasm Recurrence, Local/epidemiology , Pancreatic Neoplasms/drug therapy , Antigens, Tumor-Associated, Carbohydrate/blood , Antigens, Tumor-Associated, Carbohydrate/immunology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/immunology , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Disease-Free Survival , Drug Resistance, Neoplasm/immunology , Humans , Inhibitory Concentration 50 , Liquid Biopsy , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Polysaccharides/blood , Polysaccharides/immunology , Risk Assessment/methods
4.
Neurooncol Adv ; 2(1): vdaa067, 2020.
Article in English | MEDLINE | ID: mdl-32642717

ABSTRACT

BACKGROUND: Aberrant MET receptor tyrosine kinase (RTK) activation leads to invasive tumor growth in different types of cancer. Overexpression of MET and its ligand hepatocyte growth factor (HGF) occurs more frequently in glioblastoma (GBM) than in low-grade gliomas. Although we have shown previously that HGF-autocrine activation predicts sensitivity to MET tyrosine kinase inhibitors (TKIs) in GBM, whether it initiates tumorigenesis remains elusive. METHODS: Using a well-established Sleeping Beauty (SB) transposon strategy, we injected human HGF and MET cDNA together with a short hairpin siRNA against Trp53 (SB-hHgf.Met.ShP53) into the lateral ventricle of neonatal mice to induce spontaneous glioma initiation and characterized the tumors with H&E and immunohistochemistry analysis. Glioma sphere cells also were isolated for measuring the sensitivity to specific MET TKIs. RESULTS: Mixed injection of SB-hHgf.Met.ShP53 plasmids induced de novo glioma formation with invasive tumor growth accompanied by HGF and MET overexpression. While glioma stem cells (GSCs) are considered as the tumor-initiating cells in GBM, both SB-hHgf.Met.ShP53 tumor sections and glioma spheres harvested from these tumors expressed GSC markers nestin, GFAP, and Sox 2. Moreover, specific MET TKIs significantly inhibited tumor spheres' proliferation and MET/MAPK/AKT signaling. CONCLUSIONS: Overexpression of the HGF/MET axis along with p53 attenuation may transform neural stem cells into GSCs, resulting in GBM formation in mice. These tumors are primarily driven by the MET RTK pathway activation and are sensitive to MET TKIs. The SB-hHgf.Met.ShP53 spontaneous mouse glioma model provides a useful tool for studying GBM tumor biology and MET-targeting therapeutics.

5.
Clin Cancer Res ; 25(9): 2745-2754, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30617132

ABSTRACT

PURPOSE: The CA19-9 biomarker is elevated in a substantial group of patients with pancreatic ductal adenocarcinoma (PDAC), but not enough to be reliable for the detection or diagnosis of the disease. We hypothesized that a glycan called sTRA (sialylated tumor-related antigen) is a biomarker for PDAC that improves upon CA19-9. EXPERIMENTAL DESIGN: We examined sTRA and CA19-9 expression and secretion in panels of cell lines, patient-derived xenografts, and primary tumors. We developed candidate biomarkers from sTRA and CA19-9 in a training set of 147 plasma samples and used the panels to make case-control calls, based on predetermined thresholds, in a 50-sample validation set and a blinded, 147-sample test set. RESULTS: The sTRA glycan was produced and secreted by pancreatic tumors and models that did not produce and secrete CA19-9. Two biomarker panels improved upon CA19-9 in the training set, one optimized for specificity, which included CA19-9 and 2 versions of the sTRA assay, and another optimized for sensitivity, which included 2 sTRA assays. Both panels achieved statistical improvement (P < 0.001) over CA19-9 in the validation set, and the specificity-optimized panel achieved statistical improvement (P < 0.001) in the blinded set: 95% specificity and 54% sensitivity (75% accuracy), compared with 97%/30% (65% accuracy). Unblinding produced further improvements and revealed independent, complementary contributions from each marker. CONCLUSIONS: sTRA is a validated serological biomarker of PDAC that yields improved performance over CA19-9. The new panels may enable surveillance for PDAC among people with elevated risk, or improved differential diagnosis among patients with suspected pancreatic cancer.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/blood , Biomarkers, Tumor/blood , CA-19-9 Antigen/blood , Carcinoma, Pancreatic Ductal/diagnosis , N-Acetylneuraminic Acid/chemistry , Pancreatic Neoplasms/diagnosis , Aged , Animals , Carcinoma, Pancreatic Ductal/blood , Case-Control Studies , Female , Follow-Up Studies , Humans , Male , Mice , Middle Aged , Pancreatic Neoplasms/blood , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
J Transl Med ; 16(1): 253, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30208970

ABSTRACT

BACKGROUND: Aberrant MET tyrosine kinase signaling is known to cause cancer initiation and progression. While MET inhibitors are in clinical trials against several cancer types, the clinical efficacies are controversial and the molecular mechanisms toward sensitivity remain elusive. METHODS: With the goal to investigate the molecular basis of MET amplification (METamp) and hepatocyte growth factor (HGF) autocrine-driven tumors in response to MET tyrosine kinase inhibitors (TKI) and neutralizing antibodies, we compared cancer cells harboring METamp (MKN45 and MHCCH97H) or HGF-autocrine (JHH5 and U87) for their sensitivity and downstream biological responses to a MET-TKI (INC280) and an anti-MET monoclonal antibody (MetMab) in vitro, and for tumor inhibition in vivo. RESULTS: We find that cancer cells driven by METamp are more sensitive to INC280 than are those driven by HGF-autocrine activation. In METamp cells, INC280 induced a DNA damage response with activation of repair through the p53BP1/ATM signaling pathway. Although MetMab failed to inhibit METamp cell proliferation and tumor growth, both INC280 and MetMab reduced HGF-autocrine tumor growth. In addition, we also show that HGF stimulation promoted human HUVEC cell tube formation via the Src pathway, which was inhibited by either INC280 or MetMab. These observations suggest that in HGF-autocrine tumors, the endothelial cells are the secondary targets MET inhibitors. CONCLUSIONS: Our results demonstrate that METamp and HGF-autocrine activation favor different molecular mechanisms. While combining MET TKIs and ATM inhibitors may enhance the efficacy for treating tumors harboring METamp, a combined inhibition of MET and angiogenesis pathways may improve the therapeutic efficacy against HGF-autocrine tumors.


Subject(s)
Antibodies, Neutralizing/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , Autocrine Communication/drug effects , Benzamides , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Hepatocyte Growth Factor/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Imidazoles/pharmacology , Mice, SCID , Signal Transduction/drug effects , Triazines/pharmacology , Tumor Suppressor p53-Binding Protein 1/metabolism
7.
Anal Chem ; 89(22): 12342-12350, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29058413

ABSTRACT

Knowledge of lectin and glycosidase specificities is fundamental to the study of glycobiology. The primary specificities of such molecules can be uncovered using well-established tools, but the complex details of their specificities are difficult to determine and describe. Here we present a language and algorithm for the analysis and description of glycan motifs with high complexity. The language uses human-readable notation and wildcards, modifiers, and logical operators to define motifs of nearly any complexity. By applying the syntax to the analysis of glycan-array data, we found that the lectin AAL had higher binding where fucose groups are displayed on separate branches. The lectin SNA showed gradations in binding based on the length of the extension displaying sialic acid and on characteristics of the opposing branches. A new algorithm to evaluate changes in lectin binding upon treatment with exoglycosidases identified the primary specificities and potential fine specificities of an α1-2-fucosidase and an α2-3,6,8-neuraminidase. The fucosidase had significantly lower action where sialic acid neighbors the fucose, and the neuraminidase showed statistically lower action where α1-2 fucose neighbors the sialic acid or is on the opposing branch. The complex features identified here would have been inaccessible to analysis using previous methods. The new language and algorithms promise to facilitate the precise determination and description of lectin and glycosidase specificities.


Subject(s)
Glycoside Hydrolases/metabolism , Lectins/analysis , Microarray Analysis , Polysaccharides/chemistry , Algorithms , Binding Sites , Fucose/chemistry , Glycoside Hydrolases/analysis , Humans , Milk, Human/chemistry , Polysaccharides/chemical synthesis , Substrate Specificity
8.
Proc Natl Acad Sci U S A ; 111(7): 2590-5, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550287

ABSTRACT

A deficiency of mitogen-inducible gene-6 (Mig-6) in mice leads to the development of an early-onset, osteoarthritis (OA)-like disorder in multiple synovial joints, underlying its importance in maintaining joint homeostasis. Here we determined what joint tissues Mig-6 is expressed in and what role chondrocytes play in the Mig-6-deficient OA-like disorder. A Mig-6/lacZ reporter mouse strain expressing ß-galactosidase under the control of the Mig-6 gene promoter was generated to determine Mig-6 expression in joint tissues. By ß-galactosidase staining, we demonstrated that Mig-6 was uniquely expressed in the cells across the entire surface of the synovial joint cavity, including chondrocytes in the superficial zone of articular cartilage and in the meniscus, as well as synovial lining cells. By crossing Mig-6-floxed mice to Col2a1-Cre transgenic mice, to generate cartilage-specific deletion of Mig-6, we demonstrated that deficiency of Mig-6 in the chondrocytes results in a joint phenotype that only partially recapitulates the OA-like disorder of the Mig-6-deficient mice: Ubiquitous deletion of Mig-6 led to the OA-like disorder in multiple joints, whereas cartilage-specific deletion affected the knees but rarely other joints. Furthermore, chondrocytes with Mig-6 deficiency showed excessive proliferative activities along with enhanced EGF receptor signaling in the articular cartilage and in the abnormally formed osteophytes. Our findings provide insight into the crucial requirement for Mig-6 in maintaining joint homeostasis and in regulating chondrocyte activities in the synovial joints. Our data also suggest that other cell types are required for fully developing the Mig-6-deficient OA-like disorder.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/physiology , Intracellular Signaling Peptides and Proteins/deficiency , Osteoarthritis/genetics , Animals , Cell Proliferation , Genetic Vectors , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Osteoarthritis/pathology , Reverse Transcriptase Polymerase Chain Reaction , beta-Galactosidase
9.
Mol Cancer Ther ; 12(8): 1429-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23720767

ABSTRACT

The MET and EGFR receptor tyrosine kinases (RTK) are often coexpressed and may cross-talk in driving the development and progression of non-small cell lung carcinoma (NSCLC). In addition, MET amplification is an alternative resistance mechanism for escaping EGFR-targeted therapy. To assess the benefits of combined targeting of MET and EGFR for treating NSCLCs, we investigated the activities of these two RTK pathways in NSCLC cell lines and evaluated their responses to SGX523 and erlotinib, the small-molecule kinase inhibitors of MET and EGFR, respectively. We showed that MET interacts with and cross-activates EGFR in MET-amplified or -overexpressed cells. The inhibition of both MET and EGFR results in maximal suppression of downstream signaling and of cell proliferation when their ligands are present. Furthermore, we showed that SGX523 plus erlotinib strengthens anticancer activity in vivo in a cellular context-dependent manner. The combination led to the regression of H1993 tumors by enhancing the suppression of proliferation and inducing apoptosis, whereas H1373 tumor growth was significantly reduced by the combination via suppression of proliferation without inducing apoptosis. SGX523 alone was sufficient to achieve near-complete regression of EBC-1 tumors; its combination with erlotinib strongly inhibited the viability of a population of insensitive cells emerging from an SGX523-treated EBC-1 tumor recurrence. Our data suggest that inhibition of both MET and EGFR can enhance anticancer effects against NSCLCs in a context-dependent manner and thus provide a strong rationale for combining MET and EGFR inhibitors in treating NSCLCs.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Small Cell Lung Carcinoma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Synergism , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Humans , Lung Neoplasms/drug therapy , Mice , Molecular Targeted Therapy , Proto-Oncogene Proteins c-met/metabolism , Pyridazines/pharmacology , Quinazolines/pharmacology , Signal Transduction/drug effects , Small Cell Lung Carcinoma/drug therapy , Triazoles/pharmacology , Xenograft Model Antitumor Assays
10.
PLoS One ; 7(6): e38955, 2012.
Article in English | MEDLINE | ID: mdl-22701735

ABSTRACT

Epigenetic silencing is one of the mechanisms leading to inactivation of a tumor suppressor gene, either by DNA methylation or histone modification in a promoter regulatory region. Mitogen inducible gene 6 (MIG-6), mainly known as a negative feedback inhibitor of the epidermal growth factor receptor (EGFR) family, is a tumor suppressor gene that is associated with many human cancers. To determine if MIG-6 is inactivated by epigenetic alteration, we identified a group of human lung cancer and melanoma cell lines in which its expression is either low or undetectable and studied the effects of methylation and of histone deacetylation on its expression. The DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine (5-aza-dC) induced MIG-6 expression in melanoma cell lines but little in lung cancer lines. By contrast, the histone deacetylase (HDAC) inhibitor trichostatin A (TSA) induced MIG-6 expression in lung cancer lines but had little effect in melanoma lines. However, the MIG-6 promoter itself did not appear to be directly affected by either methylation or histone deacetylation, indicating an indirect regulatory mechanism. Luciferase reporter assays revealed that a short segment of exon 1 in the MIG-6 gene is responsible for TSA response in the lung cancer cells; thus, the MIG-6 gene can be epigenetically silenced through an indirect mechanism without having a physical alteration in its promoter. Furthermore, our data also suggest that MIG-6 gene expression is differentially regulated in lung cancer and melanoma.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Azacitidine/analogs & derivatives , DNA Modification Methylases/antagonists & inhibitors , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Azacitidine/pharmacology , Base Sequence , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , CpG Islands/genetics , DNA Primers/genetics , Decitabine , Epigenesis, Genetic/genetics , Humans , Luciferases , Microarray Analysis , Molecular Sequence Data , Plasmids/genetics , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Tumor Suppressor Proteins/genetics
11.
Cancer Res ; 70(17): 6880-90, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20643778

ABSTRACT

The hepatocyte growth factor (HGF)-MET pathway supports several hallmark cancer traits, and it is frequently activated in a broad spectrum of human cancers (http://www.vai.org/met/). With the development of many cancer drugs targeting this pathway, there is a need for relevant in vivo model systems for preclinical evaluation of drug efficacy. Here, we show that production of the human HGF ligand in transgenic severe combined immunodeficient mice (hHGF(tg)-SCID mice) enhances the growth of many MET-expressing human carcinoma xenografts, including those derived from lung, breast, kidney, colon, stomach, and pancreas. In this model, the MET-specific small-molecule kinase inhibitor SGX523 partially inhibits the HGF-dependent growth of lung, breast, and pancreatic tumors. However, much greater growth suppression is achieved by combinatorial inhibition with the epidermal growth factor receptor (EGFR) kinase inhibitor erlotinib. Together, these results validate the hHGF(tg)-SCID mouse model for in vivo determination of MET sensitivity to drug inhibition. Our findings also indicate that simultaneously targeting the MET and EGFR pathways can provide synergistic inhibitory effects for the treatment of cancers in which both pathways are activated.


Subject(s)
Carcinoma/drug therapy , ErbB Receptors/antagonists & inhibitors , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridazines/pharmacology , Quinazolines/pharmacology , Receptors, Growth Factor/antagonists & inhibitors , Triazoles/pharmacology , Animals , Carcinoma/metabolism , Carcinoma/pathology , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Drug Synergism , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Humans , Mice , Mice, Inbred C3H , Mice, SCID , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...