Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
J Appl Lab Med ; 2(5): 674-686, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-33636862

ABSTRACT

BACKGROUND: Dried blood spot (DBS) testing has been used for years in newborn screening and for other applications when obtaining blood by venipuncture is impractical or expensive. However, several technical challenges have restricted the use of DBS testing to qualitative assays or to analytes that are present in relatively high concentrations. The application of high-sensitivity detection using single molecule counting (SMC™) technology can potentially overcome the limitations of DBS as specimen source. METHODS: A method was developed for reproducibly collecting, storing, and subsequently reconstituting DBS samples to be used with assays based on the SMC technology. Before extraction, DBS samples were scanned, and the blood spot area was calculated to normalize for sample volume and spot variability. DBS sample extraction was done using an efficient high-salt extraction buffer. DBS samples were tested using SMC-based cardiac troponin I (cTnI), prostate-specific antigen (PSA), and C-reactive protein (CRP) assays. RESULTS: The SMC-DBS assays showed reproducible sensitivity, precision, and the stability required for quantifying low-abundance biomarkers. These assays were not significantly impacted by normal variations in hematocrit or sample collection technique. Correlation coefficients obtained from method comparisons between SMC-DBS and laboratory-developed tests or Food and Drug Administration-cleared tests using traditional sample types were 1.08, 1.04, and 0.99 for cTnI, PSA, and high-sensitivity CRP, respectively. CONCLUSIONS: Combining DBS finger-stick blood collection with next-generation immunoassay technology will aid the expansion of DBS testing to protein biomarkers that are in low abundance or to low-volume samples, and will enable the development and adoption of DBS testing to far-reaching applications.

2.
Breast Cancer Res Treat ; 141(1): 43-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23959396

ABSTRACT

Trastuzumab is effective in the treatment of HER2/neu over-expressing breast cancer, but not all patients benefit from it. In vitro data suggest a role for HER3 in the initiation of signaling activity involving the AKT­mTOR pathway leading to trastuzumab insensitivity. We sought to investigate the potential of HER3 alone and in the context of p95HER2 (p95), a trastuzumab resistance marker, as biomarkers of trastuzumab escape. Using the VeraTag® assay platform, we developed a dual antibody proximity-based assay for the precise quantitation of HER3 total protein (H3T) from formalin-fixed paraffin-embedded (FFPE) breast tumors. We then measured H3T in 89 patients with metastatic breast cancer treated with trastuzumab-based therapy, and correlated the results with progression-free survival and overall survival using Kaplan­Meier and decision tree analyses that also included HER2 total (H2T) and p95 expression levels. Within the sub-population of patients that over-expressed HER2, high levels of HER3 and/or p95 protein expression were significantly associated with poor clinical outcomes on trastuzumab-based therapy. Based on quantitative H3T, p95, and H2T measurements, multiple subtypes of HER2-positive breast cancer were identified that differ in their outcome following trastuzumab therapy. These data suggest that HER3 and p95 are informative biomarkers of clinical outcomes on trastuzumab therapy, and that multiple subtypes of HER2-positive breast cancer may be defined by quantitative measurements of H3T, p95, and H2T.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/secondary , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , Genes, erbB-2 , Neoplasm Proteins/biosynthesis , Receptor, ErbB-2/analysis , Receptor, ErbB-3/analysis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Breast Neoplasms/classification , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Cohort Studies , Decision Trees , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Humans , Kaplan-Meier Estimate , Neoplasm Proteins/genetics , Peptide Fragments/analysis , Peptide Fragments/immunology , Prognosis , Protein Structure, Tertiary , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptor, ErbB-3/genetics , Receptor, ErbB-3/immunology , Retrospective Studies , Single-Blind Method , Trastuzumab , Treatment Outcome
3.
PLoS One ; 6(1): e16443, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21297994

ABSTRACT

BACKGROUND: The identification of patients for targeted antineoplastic therapies requires accurate measurement of therapeutic targets and associated signaling complexes. HER3 signaling through heterodimerization is an important growth-promoting mechanism in several tumor types and may be a principal resistance mechanism by which EGFR and HER2 expressing tumors elude targeted therapies. Current methods that can study these interactions are inadequate for formalin-fixed, paraffin-embedded (FFPE) tumor samples. METHODOLOGY AND PRINCIPAL FINDINGS: Herein, we describe a panel of proximity-directed assays capable of measuring protein-interactions and phosphorylation in FFPE samples in the HER3/PI3K/Akt pathway and examine the capability of these assays to inform on the functional state of the pathway. We used FFPE breast cancer cell line and tumor models for this study. In breast cancer cell lines we observe both ligand-dependent and independent activation of the pathway and strong correlations between measured activation of key analytes. When selected cell lines are treated with HER2 inhibitors, we not only observe the expected molecular effects based on mechanism of action knowledge, but also novel effects of HER2 inhibition on key targets in the HER receptor pathway. Significantly, in a xenograft model of delayed tumor fixation, HER3 phosphorylation is unstable, while alternate measures of pathway activation, such as formation of the HER3PI3K complex is preserved. Measurements in breast tumor samples showed correlations between HER3 phosphorylation and receptor interactions, obviating the need to use phosphorylation as a surrogate for HER3 activation. SIGNIFICANCE: This assay system is capable of quantitatively measuring therapeutically relevant responses and enables molecular profiling of receptor networks in both preclinical and tumor models.


Subject(s)
Breast Neoplasms/metabolism , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Humans , Mice , Phosphorylation , Protein Binding , Receptor, ErbB-3/antagonists & inhibitors , Transplantation, Heterologous
4.
Cancer Res ; 71(5): 1871-82, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21324925

ABSTRACT

In breast cancer cells with HER2 gene amplification, HER2 receptors exist on the cell surface as monomers, homodimers, and heterodimers with EGFR/HER3. The therapeutic antibody trastuzumab, an approved therapy for HER2(+) breast cancer, cannot block ligand-induced HER2 heterodimers, suggesting it cannot effectively inhibit HER2 signaling. Hence, HER2 oligomeric states may predict the odds of a clinical response to trastuzumab in HER2-driven tumors. To test this hypothesis, we generated nontransformed human MCF10A mammary epithelial cells stably expressing a chimeric HER2-FKBP molecule that could be conditionally induced to homodimerize by adding the FKBP ligand AP1510, or instead induced to heterodimerize with EGFR or HER3 by adding the heterodimer ligands EGF/TGFα or heregulin. AP1510, EGF, and heregulin each induced growth of MCF10A cells expressing HER2-FKBP. Trastuzumab inhibited homodimer-mediated but not heterodimer-mediated cell growth. In contrast, the HER2 antibody pertuzumab, which blocks HER2 heterodimerization, inhibited growth induced by heregulin but not AP1510. Lastly, the HER2/EGFR tyrosine kinase inhibitor lapatinib blocked both homodimer- and heterodimer-induced growth. AP1510 triggered phosphorylation of Erk1/2 but not AKT, whereas trastuzumab inhibited AP1510-induced Erk1/2 phosphorylation and Shc-HER2 homodimer binding, but not TGFα-induced AKT phosphorylation. Consistent with these observations, high levels of HER2 homodimers correlated with longer time to progression following trastuzumab therapy in a cohort of patients with HER2-overexpressing breast cancer. Together, our findings confirm the notion that HER2 oligomeric states regulate HER2 signaling, also arguing that trastuzumab sensitivity of homodimers may reflect their inability to activate the PI3K (phosphoinositide 3-kinase)/AKT pathway. A clinical implication of our results is that high levels of HER2 homodimers may predict a positive response to trastuzumab.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Protein Multimerization/physiology , Receptor, ErbB-2/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Cells, Cultured , Female , Gene Expression , Gene Expression Profiling , Humans , Immunoblotting , Immunoprecipitation , Lapatinib , Middle Aged , Oligonucleotide Array Sequence Analysis , Quinazolines/pharmacology , Receptor, ErbB-2/chemistry , Signal Transduction/drug effects , Signal Transduction/physiology , Trastuzumab
5.
Diagn Mol Pathol ; 18(1): 11-21, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19214113

ABSTRACT

The availability of drugs targeting the EGFR/HER/erbB signaling pathway has created a need for diagnostics that accurately predict treatment responses. We have developed and characterized a novel assay to provide sensitive and quantitative measures of HER proteins and homodimers in formalin-fixed, paraffin-embedded (FFPE) cell lines and breast tumor tissues, to test these variables. In the VeraTag assay, HER proteins and homodimers are detected through the release of fluorescent tags conjugated to specific HER antibodies, requiring proximity to a second HER antibody. HER2 protein quantification was normalized to tumor area, and compared to receptor numbers in 12 human tumor cell lines determined by fluorescence-activated cell sorting (FACS), and with HER immunohistochemistry (IHC) test categories and histoscores in cell lines and 170 breast tumors. HER1 and HER2 expression levels determined by the VeraTag assay are proportional to receptor number over more than a 2 log10 range, and HER homodimer levels are consistent with crosslinking and immunoprecipitation results. VeraTag HER2 measurements of breast tumor tissue and cell lines correlate with standard IHC test categories (P<0.001). VeraTag HER2 levels also agree with IHC histoscores at lower HER2 protein levels, but are continuous and overlapping between IHC test categories, extending the dynamic range 5-fold to 10-fold at higher HER2 levels. The VeraTag assay specifically and reproducibly measures HER1 and HER2 protein and homodimers in FFPE tissues. The continuous measure of HER2 protein levels over a broad dynamic range, and the novel HER2 homodimer measure, are presently being assessed as predictive markers for responses to targeted HER2 therapy.


Subject(s)
Breast Neoplasms/pathology , Breast/chemistry , ErbB Receptors/analysis , Pathology, Clinical/methods , Receptor, ErbB-2/analysis , ErbB Receptors/metabolism , Female , Humans , Protein Multimerization , Receptor, ErbB-2/metabolism , Staining and Labeling/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...