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











Publication year range
2.
Breast Cancer Res ; 19(1): 91, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28784153

ABSTRACT

BACKGROUND: The effects of neoadjuvant chemotherapy on immune markers remain largely unknown. The specific aim of this study was to assess stromal tumor-infiltrating lymphocytes (TILs) and programmed death ligand 1 (PD-L1) protein expression in a cohort of breast cancer patients treated with neoadjuvant chemotherapy. METHODS: Using quantitative immunofluorescence, we investigated stromal TILs and PD-L1 protein expression in pre-treatment and residual breast cancer tissue from a Yale Cancer Center patient cohort of 58 patients diagnosed with breast cancer from 2003 to 2009 and treated with neoadjuvant chemotherapy. We compared the TIL count and PD-L1 status in paired pre-treatment and residual cancer tissues and correlated changes and baseline levels with survival. RESULTS: Of the 58 patients, 46 (79.3%) had hormone-positive and 34 (58.6%) had node-positive breast cancer. Eighty-six percent of residual cancer tissues had TIL infiltration and 17% had PD-L1 expression. There was a trend for higher TIL counts in postchemotherapy compared to prechemotherapy samples (p = 0.09). Increase in TIL count was associated with longer 5-year recurrence-free survival (p = 0.02, HR = 3.9, 95% CI = 1.179-15.39). PD-L1 expression (both stromal and tumor cells) was significantly lower in post-treatment samples (p = 0.001). Change in PD-L1 expression after therapy or TILs and PD-L1 expression in the posttreatment samples did not correlate with survival. CONCLUSIONS: Increase in stromal TILs in residual cancer compared to pretreatment tissue is associated with improved recurrence-free survival. Despite a trend for increasing TIL counts, PD-L1 expression decreased in residual disease compared to pretreatment samples.


Subject(s)
B7-H1 Antigen/genetics , Breast Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/immunology , Prognosis , Adult , Aged , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Middle Aged , Neoadjuvant Therapy/adverse effects
3.
Cancer Immunol Res ; 3(4): 326-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25527356

ABSTRACT

Programmed death 1 ligand 1 (PD-L1) is an immune regulatory molecule that limits antitumor immune activity. Targeting of PD-L1 and other immune checkpoint proteins has shown therapeutic activity in various tumor types. The expression of PD-L1 and its correlation with response to neoadjuvant chemotherapy in breast cancer has not been studied extensively. Our goal was to assess PD-L1 expression in a cohort of breast cancer patients treated with neoadjuvant chemotherapy. Pretreatment biopsies from 105 patients with breast cancer from Yale New Haven Hospital that subsequently received neoadjuvant chemotherapy were assessed for PD-L1 protein expression by automated quantitative analysis with a rabbit monoclonal antibody (E1L3N) to the cytoplasmic domain of PD-L1. In addition, tumor-infiltrating lymphocytes (TIL) were assessed on hematoxylin and eosin slides. PD-L1 expression was observed in 30% of patients, and it was positively associated with hormone-receptor-negative and triple-negative status and high levels of TILs. Both TILs and PD-L1 measured in the epithelium or stroma predicted pathologic complete response (pCR) to neoadjuvant chemotherapy in univariate and multivariate analyses. However, because they are strongly associated, TILs and PD-L1 cannot both be included in a significant multivariate model. PD-L1 expression is prevalent in breast cancer, particularly hormone-receptor-negative and triple-negative patients, indicating a subset of patients that may benefit from immune therapy. Furthermore, PD-L1 and TILs correlate with pCR, and high PD-L1 predicts pCR in multivariate analysis.


Subject(s)
B7-H1 Antigen/biosynthesis , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Aged , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Cohort Studies , Female , Humans , Middle Aged , Neoadjuvant Therapy , Neoplasm Proteins/biosynthesis , Prognosis , Treatment Outcome
4.
Clin Cancer Res ; 20(23): 5995-6005, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25255793

ABSTRACT

PURPOSE: Although tumor-infiltrating lymphocytes (TIL) have been associated with response to neoadjuvant therapy, measurement typically is subjective, semiquantitative, and unable to differentiate among subpopulations. Here, we describe a quantitative objective method for analyzing lymphocyte subpopulations and assessing their predictive value. EXPERIMENTAL DESIGN: We developed a quantitative immunofluorescence assay to measure stromal expression of CD3, CD8, and CD20 on one slide. We validated this assay by comparison with flow cytometry on tonsil specimens and assessed predictive value in breast cancer on a neoadjuvant cohort (n = 95). Then, each marker was tested for prediction of pathologic complete response (pCR) compared with pathologist estimation of the percentage of lymphocyte infiltrate. RESULTS: The lymphocyte percentage and CD3, CD8, and CD20 proportions were similar between flow cytometry and quantitative immunofluorescence on tonsil specimens. Pathologist TIL count predicted pCR [P = 0.043; OR, 4.77; 95% confidence interval (CI), 1.05-21.6] despite fair interobserver reproducibility (κ = 0.393). Stromal AQUA (automated quantitative analysis) scores for CD3 (P = 0.023; OR, 2.51; 95% CI, 1.13-5.57), CD8 (P = 0.029; OR, 2.00; 95% CI, 1.08-3.72), and CD20 (P = 0.005; OR, 1.80; 95% CI, 1.19-2.72) predicted pCR in univariate analysis. CD20 AQUA score predicted pCR (P = 0.019; OR, 5.37; 95% CI, 1.32-21.8) independently of age, size, nuclear grade, nodal status, ER, PR, HER2, and Ki-67, whereas CD3, CD8, and pathologist estimation did not. CONCLUSIONS: We have developed and validated an objective, quantitative assay measuring TILs in breast cancer. Although this work provides analytic validity, future larger studies will be required to prove clinical utility.


Subject(s)
Antigens, CD20/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , CD3 Complex/metabolism , CD8 Antigens/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Female , Humans , Immunophenotyping , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Middle Aged , Neoadjuvant Therapy , Neoplasm Grading , Neoplasm Staging , Prognosis , Reproducibility of Results , Treatment Outcome , Tumor Burden
5.
Breast Cancer Res Treat ; 146(3): 657-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25007965

ABSTRACT

Though the role of Estrogen Receptor (ER)α in breast cancer has been studied extensively, there is little consensus about the role of alternative ER isoform ERß in breast cancer biology. ERß has significant sequence homology to ERα but is located on a different chromosome and maintains both overlapping and unique functional attributes. Five variants exist, resulting from alternative splicing of the C-terminal region of ERß. The relevance of ERß variants in breast cancer outcomes and response to therapy is difficult to assess because of conflicting reports in the literature, likely due to variable methods used to assess ERß in patient tumors. Here, we quantitatively assess expression of ERß splice variants on over 2,000 breast cancer patient samples. Antibodies against ERß variants were validated for staining specificity in cell lines by siRNA knockdown of ESR2 and staining reproducibility on formalin-fixed paraffin-embedded tissue by quantitative immunofluorescence (QIF) using AQUA technology. We found antibodies against splice variants ERß1 and ERß5, but not ERß2/cx, which were sensitive, specific, and reproducible. QIF staining of validated antibodies showed both ERß1 and ERß5 QIF scores, which have a normal (bell shaped) distribution on most cohorts assessed, and their expression is significantly associated with each other. Extensive survival analyses show that ERß1 is not a prognostic or predictive biomarker for breast cancer. ERß5 appears to be a context-dependent marker of worse outcome in HER2-positive and triple-negative patients, suggesting an unknown biological function in the absence of ERα.


Subject(s)
Estrogen Receptor beta/biosynthesis , Prognosis , Protein Isoforms/biosynthesis , Triple Negative Breast Neoplasms/genetics , Adult , Aged , Alternative Splicing/genetics , Cohort Studies , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , Middle Aged , Protein Isoforms/genetics , RNA, Small Interfering , Triple Negative Breast Neoplasms/pathology
6.
Clin Cancer Res ; 20(10): 2773-82, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24647569

ABSTRACT

PURPOSE: Blockade of the PD-1/PD-L1 axis emerged as a promising new therapeutic option for cancer that has resulted in lasting responses in metastatic renal, lung carcinomas, and melanomas. Tumor PD-L1 protein expression may predict response to drugs targeting this pathway. Measurement of PD-L1 protein is limited by the lack of standardized immunohistochemical methods and variable performance of antibodies. Our goal was to correlate PD-L1 mRNA expression with clinical variables in primary breast carcinomas. EXPERIMENTAL DESIGN: The fluorescent RNAscope paired-primer assay was used to quantify in situ PD-L1 mRNA levels in 636 stage I-III breast carcinomas on two sets of tissue microarrays [YTMA128 (n = 238) and YTMA201 (n = 398)]. Tumor-infiltrating lymphocytes (TIL) were assessed by hematoxylin/eosin stain and quantitative fluorescence. RESULTS: On YTMA128 and YTMA201, 55.7% and 59.5% of cases showed PD-L1 mRNA expression, respectively. Higher PD-L1 mRNA expression was significantly associated with increased TILs (P = 0.04) but not with other clinical variables. Elevated TILs (scores 2 and 3+) occurred in 16.5% on YTMA128 and 14.8% on YTMA201 and was associated with estrogen receptor-negative status (P = 0.01 on YTMA128 and 0.0001 on YTMA201). PD-L1 mRNA expression was associated with longer recurrence-free survival (log-rank P = 0.01), which remained significant in multivariate analysis including age, tumor size, histologic grade, nodal metastasis, hormone receptor, HER2 status, and the extent of TILs (HR, 0.268; CI, 0.099-0.721; P = 0.009). CONCLUSIONS: PD-L1 mRNA expression is identified in nearly 60% of breast tumors and it is associated with increased TILs and improved recurrence-free survival. These observations support the evaluation of PD-1/PD-L1-targeted therapies in breast cancer.


Subject(s)
B7-H1 Antigen/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Lymphocytes, Tumor-Infiltrating/metabolism , B7-H1 Antigen/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Fluorescent Antibody Technique/methods , Humans , In Situ Hybridization , Kaplan-Meier Estimate , Lymphatic Metastasis , Lymphocytes, Tumor-Infiltrating/pathology , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Tissue Array Analysis
7.
Nat Commun ; 4: 2115, 2013.
Article in English | MEDLINE | ID: mdl-23828459

ABSTRACT

Double-stranded DNA ends, often from replication, drive genomic instability, yet their origin in non-replicating cells is unknown. Here we show that transcriptional RNA/DNA hybrids (R-loops) generate DNA ends that underlie stress-induced mutation and amplification. Depleting RNA/DNA hybrids with overproduced RNase HI reduces both genomic changes, indicating RNA/DNA hybrids as intermediates in both. An Mfd requirement and inhibition by translation implicate transcriptional R-loops. R-loops promote instability by generating DNA ends, shown by their dispensability when ends are provided by I-SceI endonuclease. Both R-loops and single-stranded endonuclease TraI are required for end formation, visualized as foci of a fluorescent end-binding protein. The data suggest that R-loops prime replication forks that collapse at single-stranded nicks, producing ends that instigate genomic instability. The results illuminate how DNA ends form in non-replicating cells, identify R-loops as the earliest known mutation/amplification intermediate, and suggest that genomic instability during stress could be targeted to transcribed regions, accelerating adaptation.


Subject(s)
DNA Damage , Escherichia coli/growth & development , Escherichia coli/genetics , Genomic Instability/genetics , Nucleic Acid Heteroduplexes/metabolism , Bacterial Proteins/metabolism , DNA Breaks, Double-Stranded , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , Models, Genetic , Mutagenesis/genetics , Point Mutation/genetics , Protein Biosynthesis , Ribonucleases/metabolism , Stress, Physiological/genetics , Transcription Factors/metabolism , Transcription, Genetic , Viral Proteins/metabolism
8.
Appl Immunohistochem Mol Morphol ; 21(2): 139-47, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22820659

ABSTRACT

Studies comparing rabbit monoclonal SP1 antibody with 1D5 for estrogen receptor (ER) immunohistochemical testing show conflicting results. Here we use a standardized quantitative immunofluorescent (QIF) ER assay to determine the level and significance of discordance between the antibodies. Both antibodies were assessed by QIF on our Index TMA of cell lines and case controls, followed by QIF and immunohistochemical analysis on 2 retrospective cohorts from Yale. On the Index TMA, SP1 displayed stronger signal-to-noise ratio compared with 1D5. On the patient cohorts, the range of discrepancy between the 2 antibodies was 8% to 16.9%, with the majority of discrepant cases being SP1 positive/1D5 negative. Kaplan-Meier analysis of the discrepant cases showed outcomes comparable to those of double-positive cases, suggesting that SP1 is more sensitive than 1D5. A series of cases with high levels of ER-ß shows that neither antibody cross-reacts, suggesting equivalent specificity. Future efforts are needed to determine whether response to endocrine therapies show superiority of either antibody as a companion diagnostic test.


Subject(s)
Antibodies, Monoclonal/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Estrogen Receptor beta/genetics , Fluorescent Antibody Technique, Direct/standards , Adult , Animals , Antibodies, Monoclonal/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , False Negative Reactions , Gene Expression , Humans , Kaplan-Meier Estimate , Middle Aged , Rabbits , Retrospective Studies , Signal-To-Noise Ratio , Tissue Array Analysis
9.
Ann N Y Acad Sci ; 1267: 103-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22954223

ABSTRACT

Gross chromosomal rearrangements (GCRs), or changes in chromosome structure, play central roles in evolution and are central to cancer formation and progression. GCRs underlie copy number variation (CNV), and therefore genomic disorders that stem from CNV. We study amplification in Escherichia coli as a model system to understand mechanisms and circumstances of GCR formation. Here, we summarize observations that led us to postulate that GCR occurs by a replicative mechanism as part of activated stress responses. We report that we do not find RecA to be downregulated by stress on a population basis and that constitutive expression of RecA does not inhibit amplification, as would be expected if downregulation of RecA made cells permissive for nonhomologous recombination. Strains deleted for the genes for three proteins that inhibit RecA activity, psiB, dinI, and recX, all show unaltered amplification, suggesting that if they do downregulate RecA indirectly, this activity does not promote amplification.


Subject(s)
Chromosomes, Bacterial/genetics , DNA Replication , Escherichia coli/genetics , Stress, Physiological/genetics , DNA Copy Number Variations , DNA End-Joining Repair , DNA, Bacterial/genetics , Escherichia coli/physiology , Gene Amplification , Gene Rearrangement , Point Mutation
10.
Biotechniques ; 52(4): 235-45, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22482439

ABSTRACT

MicroRNAs (miRNAs) have emerged as key regulators in the pathogenesis of cancers where they can act as either oncogenes or tumor suppressors. Most miRNA measurement methods require total RNA extracts which lack critical spatial information and present challenges for standardization. We have developed and validated a method for the quantitative analysis of miRNA expression by in situ hybridization (ISH) allowing for the direct assessment of tumor epithelial expression of miRNAs. This co-localization based approach (called qISH) utilizes DAPI and cytokeratin immunofluorescence to establish subcellular compartments in the tumor epithelia, then multiplexed with the miRNA ISH, allows for quantitative measurement of miRNA expression within these compartments. We use this approach to assess miR-21, miR-92a, miR-34a, and miR-221 expression in 473 breast cancer specimens on tissue microarrays. We found that miR-221 levels are prognostic in breast cancer illustrating the high-throughput method and confirming that miRNAs can be valuable biomarkers in cancer. Furthermore, in applying this method we found that the inverse relationship between miRNAs and proposed target proteins is difficult to discern in large population cohorts. Our method demonstrates an approach for large cohort, tissue microarray-based assessment of miRNA expression.


Subject(s)
In Situ Hybridization/methods , MicroRNAs/metabolism , Tissue Array Analysis/methods , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , Mice , Mice, Knockout , MicroRNAs/genetics , Neoplasm Proteins/metabolism , Prognosis , Reproducibility of Results
11.
Cancer Cell ; 20(1): 119-31, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21741601

ABSTRACT

Endocrine therapies for breast cancer that target the estrogen receptor (ER) are ineffective in the 25%-30% of cases that are ER negative (ER-). Androgen receptor (AR) is expressed in 60%-70% of breast tumors, independent of ER status. How androgens and AR regulate breast cancer growth remains largely unknown. We find that AR is enriched in ER- breast tumors that overexpress HER2. Through analysis of the AR cistrome and androgen-regulated gene expression in ER-/HER2+ breast cancers we find that AR mediates ligand-dependent activation of Wnt and HER2 signaling pathways through direct transcriptional induction of WNT7B and HER3. Specific targeting of AR, Wnt or HER2 signaling impairs androgen-stimulated tumor cell growth suggesting potential therapeutic approaches for ER-/HER2+ breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Androgens/pharmacology , Anilides/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dihydrotestosterone/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Mice , Nitriles/pharmacology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tosyl Compounds/pharmacology , Transcriptional Activation/drug effects , Up-Regulation/drug effects , Wnt Proteins/genetics , Wnt Proteins/metabolism , Xenograft Model Antitumor Assays , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL