Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Hepatology ; 77(3): 774-788, 2023 03 01.
Article in English | MEDLINE | ID: mdl-35908246

ABSTRACT

BACKGROUND AND AIMS: The sensitivity of current surveillance methods for detecting early-stage hepatocellular carcinoma (HCC) is suboptimal. Extracellular vesicles (EVs) are promising circulating biomarkers for early cancer detection. In this study, we aim to develop an HCC EV-based surface protein assay for early detection of HCC. APPROACH AND RESULTS: Tissue microarray was used to evaluate four potential HCC-associated protein markers. An HCC EV surface protein assay, composed of covalent chemistry-mediated HCC EV purification and real-time immuno-polymerase chain reaction readouts, was developed and optimized for quantifying subpopulations of EVs. An HCC EV ECG score, calculated from the readouts of three HCC EV subpopulations ( E pCAM + CD63 + , C D147 + CD63 + , and G PC3 + CD63 + HCC EVs), was established for detecting early-stage HCC. A phase 2 biomarker study was conducted to evaluate the performance of ECG score in a training cohort ( n  = 106) and an independent validation cohort ( n  = 72).Overall, 99.7% of tissue microarray stained positive for at least one of the four HCC-associated protein markers (EpCAM, CD147, GPC3, and ASGPR1) that were subsequently validated in HCC EVs. In the training cohort, HCC EV ECG score demonstrated an area under the receiver operating curve (AUROC) of 0.95 (95% confidence interval [CI], 0.90-0.99) for distinguishing early-stage HCC from cirrhosis with a sensitivity of 91% and a specificity of 90%. The AUROCs of the HCC EV ECG score remained excellent in the validation cohort (0.93; 95% CI, 0.87-0.99) and in the subgroups by etiology (viral: 0.95; 95% CI, 0.90-1.00; nonviral: 0.94; 95% CI, 0.88-0.99). CONCLUSION: HCC EV ECG score demonstrated great potential for detecting early-stage HCC. It could augment current surveillance methods and improve patients' outcomes.


Subject(s)
Carcinoma, Hepatocellular , Extracellular Vesicles , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Biomarkers, Tumor/analysis , Extracellular Vesicles/chemistry , Membrane Proteins , Electrocardiography , Glypicans
2.
Nature ; 510(7505): 397-401, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24828042

ABSTRACT

Metabolism and ageing are intimately linked. Compared with ad libitum feeding, dietary restriction consistently extends lifespan and delays age-related diseases in evolutionarily diverse organisms. Similar conditions of nutrient limitation and genetic or pharmacological perturbations of nutrient or energy metabolism also have longevity benefits. Recently, several metabolites have been identified that modulate ageing; however, the molecular mechanisms underlying this are largely undefined. Here we show that α-ketoglutarate (α-KG), a tricarboxylic acid cycle intermediate, extends the lifespan of adult Caenorhabditis elegans. ATP synthase subunit ß is identified as a novel binding protein of α-KG using a small-molecule target identification strategy termed drug affinity responsive target stability (DARTS). The ATP synthase, also known as complex V of the mitochondrial electron transport chain, is the main cellular energy-generating machinery and is highly conserved throughout evolution. Although complete loss of mitochondrial function is detrimental, partial suppression of the electron transport chain has been shown to extend C. elegans lifespan. We show that α-KG inhibits ATP synthase and, similar to ATP synthase knockdown, inhibition by α-KG leads to reduced ATP content, decreased oxygen consumption, and increased autophagy in both C. elegans and mammalian cells. We provide evidence that the lifespan increase by α-KG requires ATP synthase subunit ß and is dependent on target of rapamycin (TOR) downstream. Endogenous α-KG levels are increased on starvation and α-KG does not extend the lifespan of dietary-restricted animals, indicating that α-KG is a key metabolite that mediates longevity by dietary restriction. Our analyses uncover new molecular links between a common metabolite, a universal cellular energy generator and dietary restriction in the regulation of organismal lifespan, thus suggesting new strategies for the prevention and treatment of ageing and age-related diseases.


Subject(s)
Caenorhabditis elegans/drug effects , Ketoglutaric Acids/pharmacology , Longevity/physiology , Mitochondrial Proton-Translocating ATPases/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , HEK293 Cells , Humans , Jurkat Cells , Longevity/drug effects , Longevity/genetics , Mice , Mitochondrial Proton-Translocating ATPases/genetics , Protein Binding
3.
Carcinogenesis ; 39(7): 860-868, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29897425

ABSTRACT

Increasingly affordable high-throughput molecular profiling technologies have made feasible the measurement of omics-wide interindividual variations for the purposes of predicting cancer prognosis. While multiple types of genetic, epigenetic and expression changes have been implicated in ovarian cancer, existing prognostic biomarker strategies are constrained to analyzing a single class of molecular variations. The extra predictive power afforded by the integration of multiple omics types remains largely unexplored. In this study, we performed integrative analysis on tumor-based exome-, transcriptome- and methylome-wide molecular profiles from The Cancer Genome Atlas (TCGA) for variations in cancer-relevant genes to construct robust, cross-validated multiomic predictors for ovarian cancer survival. These integrated polygenic survival scores (PSSs) were able to predict 5-year overall (OS) and progression-free survival in the Caucasian subsample with high accuracy (AUROC = 0.87 and 0.81, respectively). These findings suggest that the PSSs are able to predict long-term OS in TCGA patients with accuracy beyond that of previously proposed protein-based biomarker strategies. Our findings reveal the promise of an integrated omics-based approach in enhancing existing prognostic strategies. Future investigations should be aimed toward prospective external validation, strategies for standardizing application and the integration of germline variants.


Subject(s)
Ovarian Neoplasms/genetics , Biomarkers, Tumor/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Exome/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Genetic Variation/genetics , Genome, Human/genetics , Humans , Middle Aged , Prognosis , Progression-Free Survival , Transcriptome/genetics
4.
Cancers (Basel) ; 16(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38672563

ABSTRACT

Breast cancer (BC) remains among the most commonly diagnosed cancers in women worldwide. Triple-negative BC (TNBC) is a subset of BC characterized by aggressive behavior, a high risk of distant recurrence, and poor overall survival rates. Chemotherapy is the backbone for treatment in patients with TNBC, but outcomes remain poor compared to other BC subtypes, in part due to the lack of recognized functional targets. In this study, the expression of the tetraspan protein epithelial membrane protein 2 (EMP2) was explored as a predictor of TNBC response to standard chemotherapy. We demonstrate that EMP2 functions as a prognostic biomarker for patients treated with taxane-based chemotherapy, with high expression at both transcriptomic and protein levels following treatment correlating with poor overall survival. Moreover, we show that targeting EMP2 in combination with docetaxel reduces tumor load in syngeneic and xenograft models of TNBC. These results provide support for the prognostic and therapeutic potential of this tetraspan protein, suggesting that anti-EMP2 therapy may be beneficial for the treatment of select chemotherapy-resistant TNBC tumors.

5.
Cancer Res Commun ; 3(8): 1628-1637, 2023 08.
Article in English | MEDLINE | ID: mdl-37621406

ABSTRACT

Purpose: Neoadjuvant combination immune checkpoint blockade and intralesional oncolytic virotherapy have the potential to activate antitumor responses in patients with breast cancer. Experimental Design: Eligibility for this pilot phase I trial included patients with localized HER2-negative breast cancer who received systemic nivolumab and ipilimumab and intratumor talimogene laherparepvec (T-VEC; NCT04185311). The primary objective was to evaluate the safety and adverse event profile of immunotherapy combined with T-VEC in patients with localized, HER2-negative breast cancer. Results: Six patients were enrolled, 4 having relapses after prior neoadjuvant chemotherapy and 2 who were previously untreated. Toxicities included 1 patient having grade 3 hypotension and type 1 diabetes mellitus, 3 patients with hypothyroidism, and all patients having constitutional symptoms known to be associated with the administration of T-VEC. One patient had a pathologic complete response, 3 patients had pathologic partial responses, 1 showed no significant response, and 1 had disease progression. Biopsies demonstrated increased immune cell infiltration in samples from patients who responded to therapy. Conclusions: This triple immunotherapy regimen provided responses in patients with advanced or relapsed HER2-negative breast cancer, at the expense of long-term toxicities. Significance: Systemic immune checkpoint blockade with a programmed death receptor 1 and a CTL antigen-4 blocking antibody, combined with intralesional oncolytic virotherapy, is a chemotherapy-free combination aimed at inducing an antitumor immune response locally and systemic immunity.


Subject(s)
Breast Neoplasms , Melanoma , Oncolytic Virotherapy , Female , Humans , Breast Neoplasms/therapy , Immune Checkpoint Inhibitors , Ipilimumab/therapeutic use , Neoadjuvant Therapy , Neoplasm Recurrence, Local , Nivolumab/therapeutic use , Oncolytic Virotherapy/adverse effects , Pilot Projects
6.
J Proteome Res ; 11(10): 5034-45, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22934887

ABSTRACT

We have begun an early phase of biomarker discovery in three clinically important types of breast cancer using a panel of human cell lines: HER2 positive, hormone receptor positive and HER2 negative, and triple negative (HER2-, ER-, PR-). We identified and characterized the most abundant secreted, sloughed, or leaked proteins released into serum free media from these breast cancer cell lines using a combination of protein fractionation methods before LC-MS/MS mass spectrometry analysis. A total of 249 proteins were detected in the proximal fluid of 7 breast cancer cell lines. The expression of a selected group of high abundance and/or breast cancer-specific potential biomarkers including thromobospondin 1, galectin-3 binding protein, cathepsin D, vimentin, zinc-α2-glycoprotein, CD44, and EGFR from the breast cancer cell lines and in their culture media were further validated by Western blot analysis. Interestingly, mass spectrometry identified a cathepsin D protein single-nucleotide polymorphism (SNP) by alanine to valine replacement from the MCF-7 breast cancer cell line. Comparison of each cell line media proteome displayed unique and consistent biosignatures regardless of the individual group classifications, demonstrating the potential for stratification of breast cancer. On the basis of the cell line media proteome, predictive Tree software was able to categorize each cell line as HER2 positive, HER2 negative, and hormone receptor positive and triple negative based on only two proteins, muscle fructose 1,6-bisphosphate aldolase and keratin 19. In addition, the predictive Tree software clearly identified MCF-7 cell line overexpresing the HER2 receptor with the SNP cathepsin D biomarker.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Proteome/metabolism , Amino Acid Sequence , Analysis of Variance , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/isolation & purification , Breast Neoplasms/pathology , Cathepsin D/chemistry , Cathepsin D/genetics , Cathepsin D/metabolism , Chromatography, Liquid , Culture Media, Conditioned/chemistry , Female , Fructose-Bisphosphate Aldolase/metabolism , Humans , Keratin-19/metabolism , MCF-7 Cells , Molecular Sequence Data , Peptide Fragments/chemistry , Polymorphism, Single Nucleotide , Principal Component Analysis , Proteome/chemistry , Proteome/isolation & purification , Proteomics , Receptor, ErbB-2/metabolism , Tandem Mass Spectrometry
7.
Clin Proteomics ; 6(3): 93-103, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20930921

ABSTRACT

INTRODUCTION: It is widely believed that discovery of specific, sensitive, and reliable tumor biomarkers can improve the treatment of cancer. Currently, there are no obvious targets that can be used in treating triple-negative breast cancer (TNBC). METHODS: To better understand TNBC and find potential biomarkers for targeted treatment, we combined a novel hydrophobic fractionation protocol with mass spectrometry LTQ-orbitrap to explore and compare the hydrophobic sub-proteome of TNBC with another subtype of breast cancer, hormone-receptor-positive-Her2-negative breast cancer (non-TNBC). RESULTS: Hydrophobic sub-proteome of breast cancer is rich in membrane proteins. Hundreds of proteins with various defined key cellular functions were identified from TNBC and non-TNBC tumors. In this study, protein profiles of TNBC and non-TNBC were systematically examined, compared, and validated. We have found that nine keratins are down-regulated and several heat shock proteins are up-regulated in TNBC tissues. Our study may provide insights of molecules that are responsible for the aggressiveness of TNBC. CONCLUSION: The initial results obtained using a combination of hydrophobic fractionation and nano-LC mass spectrometry analysis of these proteins appear promising in the discovery of potential cancer biomarkers and bio-signatures. When sufficiently refined, this approach may prove useful in improving breast cancer treatment.

8.
Ann Diagn Pathol ; 14(4): 260-3, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20637431

ABSTRACT

Breast cancer is the leading cause of cancer in women and the third leading cause of cancer mortality in the United States. We report a case of a patient who underwent bilateral simple mastectomies and right sentinel node biopsy for invasive lobular carcinoma in the right breast. An ipsilateral sentinel lymph node showed a microscopic focus of ductal elements. Although residual lobular carcinoma was identified in the right breast, no ductal carcinoma was identified in either breast. The ducts were discrete distributed in a 3-mm focus in the lymph node parenchyma as well as the subcapsular sinus. By immunohistochemistry, the ducts were positive for cytokeratin, estrogen receptor/progesterone receptor and did not show a myoepithelial layer by P63 or smooth-muscle myosin heavy-chain staining. The differential diagnosis includes heterotopic epithelial inclusions and benign mechanical transport. Mechanical transport of the breast tissue was ruled out because primary tumor type in the breast and the ductal structures in the lymph nodes were of different types. The diagnosis of occult metastatic tumor was based on the lack of the myoepithelial layers associated with the ductal structures. The diagnostic dilemma of the differential diagnoses is discussed, and pertinent literature is reviewed.


Subject(s)
Breast , Carcinoma, Ductal, Breast/secondary , Choristoma , Lymph Nodes/pathology , Neoplasms, Unknown Primary/pathology , Breast Neoplasms/pathology , Carcinoma, Lobular/pathology , Diagnosis, Differential , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Middle Aged , Neoplasms, Multiple Primary/pathology
9.
Int J Oncol ; 30(1): 145-54, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17143523

ABSTRACT

Screening mammography is the most effective tool available for breast cancer detection. While screening mammography saves lives, it has intrinsic problems that limit further improvement. We hypothesize that protein biomarkers in nipple aspirate fluid (NAF) may separate the cancer from the non-cancer state, and therefore can be used for breast cancer detection. In this study the proteins in NAF were analyzed by surface-enhanced laser desorption ionization coupled to time-of-flight mass spectrometry (SELDI-TOF) in the m/z 5,000-85,000 range. Two methods were used to normalize spectra. Then differentially expressed signals that separate cancer from non-cancer conditions were selected by two specifically developed statistical algorithms. Proteins of interest were identified by combined liquid chromatography-tandem mass spectrometry. A set of 8 markers were identified which collectively gave 63% sensitivity, 89% specificity and 76% accuracy for distinguishing cancer from non-cancer. Further improvements in the specificity and sensitivity of this strategy could come from the development of methods for more precise quantification of the biomarkers of interest and also from focusing on the low abundant components that are not evident when unfractionated NAF is analyzed directly.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Nipples/pathology , Biopsy, Needle , Chromatography, Liquid , Cytochromes c/analysis , Female , Humans , Mass Spectrometry , Multivariate Analysis , Myoglobin/analysis , Sensitivity and Specificity
10.
Oncotarget ; 8(24): 38294-38308, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28418843

ABSTRACT

Breast cancer (BC) is a leading cause of cancer-related death in women. Adjuvant systemic chemotherapies are effective in reducing risks of recurrence and have contributed to reduced BC mortality. Although targeted adjuvant treatments determined by biomarkers for endocrine and HER2-directed therapies are largely successful, predicting clinical benefit from chemotherapy is more challenging. Drug resistance is a major reason for treatment failures. Efforts are ongoing to find biomarkers to select patients most likely to benefit from chemotherapy. Importantly, cell surface biomarkers CD44+/CD24- are linked to drug resistance in some reports, yet underlying mechanisms are largely unknown. This study focused on the potential role of CD24 expression in resistance to either docetaxel or doxorubicin in part by the use of triple-negative BC (TNBC) tissue microarrays. In vitro assays were also done to assess changes in CD24 expression and differential drug susceptibility after chemotherapy. Further, mouse tumor xenograft studies were done to confirm in vitro findings. Overall, the results show that patients with CD24-positive TNBC had significantly worse overall survival and disease-free survival after taxane-based treatment. Also, in vitro cell studies show that CD44+/CD24+/high cells are more resistant to docetaxel, while CD44+/CD24-/low cells are resistant to doxorubicin. Both in vitro and in vivo studies show that cells with CD24-knockdown are more sensitive to docetaxel, while CD24-overexpressing cells are more sensitive to doxorubicin. Further, mechanistic studies indicate that Bcl-2 and TGF-ßR1 signaling via ATM-NDRG2 pathways regulate CD24. Hence, CD24 may be a biomarker to select chemotherapeutics and a target to overcome TNBC drug resistance.


Subject(s)
CD24 Antigen/biosynthesis , Drug Resistance, Neoplasm , Triple Negative Breast Neoplasms , Animals , Biomarkers, Tumor/analysis , Cell Line, Tumor , Disease-Free Survival , Female , Heterografts , Humans , Kaplan-Meier Estimate , Mice , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology
11.
Hum Pathol ; 37(12): 1583-91, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16949910

ABSTRACT

Annexin A1 (ANXA1) is a calcium- and phospholipid-binding protein and a known mediator of glucocorticoid-regulated inflammatory responses. Using a combined multiple high-throughput approach, we recently identified a reduced expression of ANXA1 in human breast cancer. The finding was confirmed at the gene level by quantitative reverse transcription-polymerase chain reaction and at the protein level by immunohistochemical staining of normal, benign, and malignant breast tissues. In this study, we constructed and used a high-density human breast cancer tissue microarray to characterize the expressional pattern of ANXA1 according to histopathologies. The tissue microarray contains 1,158 informative breast tissue cores of different histologies including normal tissues, hyperplasia, in situ and invasive tumors, and lymph node metastases. Our results showed that there was a significant decrease in glandular expression of ANXA1 in ductal carcinoma in situ and invasive ductal carcinoma compared with either normal breast tissue or hyperplasia (P < .0001). Moreover, in benign breast tissue, myoepithelial cells showed strong expression of ANXA1. There was a decrease of ANXA1 expression in myoepithelial cells in ductal carcinoma in situ lesions compared with the same cell population in either normal or hyperplastic lesions. These results suggest that suppressed ANXA1 expression in breast tissue is correlated with breast cancer development and progression.


Subject(s)
Annexin A1/biosynthesis , Breast Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/pathology , Disease Progression , Humans , Hyperplasia/metabolism , Immunohistochemistry , Lymphatic Metastasis/pathology , Reverse Transcriptase Polymerase Chain Reaction
12.
Breast ; 15(2): 167-72, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16095904

ABSTRACT

Core biopsies are commonly used in the diagnosis of breast cancer and often are the only sample for providing prognostic and predictive markers prior to neoadjuvant chemotherapy. We retrospectively studied 87 patients with breast cancer to compare the concordance rates for tumor type, grade, estrogen receptor/progesterone receptor (ER/PR), p53 status and Her2/neu by immunohistochemistry (IHC) and fluorescent in situ hybridization (FISH) between core and excisional biopsy specimens. The histologic type of cancer had a 100% concordance rate between core and excisional biopsy specimens. The concordance rate of modified Bloom-Richardson score between core and excisional biopsy specimens was 77%, ER was 95%, PR was 89%, and p53 was 86%. The concordance rate for Her2/neu by IHC was 96% and that for FISH was 100% between the core and excisional biopsy specimens. Although breast cancer may have heterogeneous histological and immunohistochemical findings, our study shows that relatively high concordance rates can be obtained when comparing core and excisional biopsy specimens.


Subject(s)
Biomarkers, Tumor , Biopsy/standards , Breast Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Medical Records , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Receptors, Estrogen , Receptors, Progesterone , Retrospective Studies
13.
Cancer Res ; 63(6): 1280-7, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12649188

ABSTRACT

Due to the high incidence of breast cancer and associated mortality rate,the development of an effective vaccine may be beneficial for the prevention or adjuvant treatment of this malignancy. We have constructed a novel breast cancer vaccine, Bacillus Calmette-Guérin (BCG)-hIL2MUC1, that consists of BCG and expresses a truncated form of MUC1 and human interleukin (IL)-2. In vitro analysis of the BCG-hIL2MUC1 construct confirmed coexpression of MUC1 and human IL-2. The ability of BCG-hIL2MUC1 to inhibit breast cancer growth was evaluated in hu-PBL-SCID mice (severe combined immunodeficient mice reconstituted with 50 x 10(6) human peripheral blood lymphocytes) that received three biweekly injections of BCG-hIL2MUC1 (0.5 colony-forming unit). Control animals received PBS, MUC1 peptide (100 microg), or empty vector BCG-261 (0.5 colony-forming unit) vaccination. After immunization, hu-PBL-SCID mice (n = 8 in each group) were xenografted with 4 x 10(6) ZR75-1 human breast cancer cells. Whereas mice receiving the control vaccines developed a tumor, only 87% of BCG-hIL2MUC1-immunized animals developed a palpable tumor with a slower rate of tumor growth (P < 0.001). Histological analysis of the primary tumors in BCG-hIL2MUC1-immunized animals revealed areas of reduced MUC1 expression. CD8-positive human lymphocytes were detected only in tumors grown in BCG-hIL2MUC1-immunized animals. These results imply a critical role of coexpressed IL-2 and MUC1 in eliciting tumor-specific immune response. To our knowledge, this is the first report of BCG engineered to express a tumor-associated antigen. Our results suggest that BCG-hIL2MUC1 immunization inhibited breast cancer growth in hu-PBL-SCID mice. Therefore, BCG-hIL2MUC1 may be a promising candidate as a breast cancer vaccine.


Subject(s)
BCG Vaccine/pharmacology , Breast Neoplasms/therapy , Cancer Vaccines/pharmacology , Interleukin-2/immunology , Mucin-1/immunology , Amino Acid Sequence , Animals , BCG Vaccine/genetics , BCG Vaccine/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Female , Humans , Immunity, Cellular/immunology , Interleukin-2/genetics , Mice , Mice, SCID , Molecular Sequence Data , Mucin-1/genetics , Tumor Cells, Cultured , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/pharmacology , Xenograft Model Antitumor Assays
14.
Oncotarget ; 7(49): 80655-80663, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27811364

ABSTRACT

E-cadherin (CDH1) is a glycoprotein that mediates adhesion between epithelial cells and also suppresses cancer invasion. Mutation or deletion of the CDH1 gene has been reported in 30-60% cases of invasive lobular carcinoma (ILC). However, little is known about genomic differences between ILC with and without a CDH1 alteration. Therefore, we analyzed whole genome sequencing data of 169 ILC cases from The Cancer Genome Atlas (TCGA) to address this deficiency. Our study shows that CDH1 gene was altered in 59.2% (100/169) of ILC. No significant difference was identified between CDH1-altered and -unaltered ILC cases for any of the examined demographic, clinical or pathologic characteristics, including histologic grade, tumor stage, lymph node metastases, or ER/PR/HER2 states. Seven recurrent mutations (PTEN, MUC16, ERBB2, FAT4, PCDHGA2, HERC1 and FLNC) and four chromosomal changes with recurrent copy number variation (CNV) (11q13, 17q12-21, 8p11 and 8q11) were found in ILC, which correlated with a positive or negative CDH1 alteration status, respectively. The prevalence of the most common breast cancer driver abnormalities including TP53 and PIK3CA mutations and MYC and ERBB2 amplifications showed no difference between the two groups. However, CDH1-altered ILC with an ERBB2 mutation shows a significantly worse prognosis compared to its counterparts without such a mutation. Our study suggests that CDH1-altered ILC patients with ERBB2 mutations may represent an actionable group of patients who could benefit from targeted breast cancer therapy.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Cadherins/genetics , Carcinoma, Ductal, Breast/genetics , Carcinoma, Lobular/genetics , Mutation , Receptor, ErbB-2/genetics , Antigens, CD , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/therapy , Carcinoma, Lobular/enzymology , Carcinoma, Lobular/pathology , Carcinoma, Lobular/therapy , Computational Biology , DNA Mutational Analysis , Databases, Genetic , Disease-Free Survival , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Neoplasm Invasiveness , Phenotype , Precision Medicine , Risk Factors , Survival Analysis , Time Factors
15.
Int J Mol Med ; 15(2): 205-12, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15647832

ABSTRACT

Publicly available human genomic sequence data provide an unprecedented opportunity for researchers to decode the functionality of human genome. Such information is extremely valuable in cancer prevention diagnosis and treatment. Cancer Genome Anatomy Project (CGAP) and Gene Expression Omnibus (GEO) are two bioinformatic infrastructures for studying functional genomics. The goal of this study is to explore the feasibility of incorporating the Internet-available bioinformatic databases to discover human breast cancer-related genes. Several tools including the Gene Finder, Virtual Northern (vNorthern) and SAGE digital gene expression displayer (DGED) were used to analyze differential gene expression between benign and malignant breast tissues. A pilot study was performed using both EST and SAGE vNorthern to analyze the expression of a panel of known genes, including high abundance genes beta-actin and G3PDH, low abundance genes BRCA1 and p53, tissue-specific genes CEA and PSA and two breast cancer-related genes Her2/neu and MUC1. We found a high expression of beta-actin and G3PDH and a low expression of BRCA1 and p53 across different types of tissues as well as a tissue-specific expression of CEA in colon and PSA in prostate. A further analysis of 30 known breast cancer-related genes in breast cancer tissues by vNorthern demonstrated a high expression of oncogenes and low expression of tumor suppressor genes. An open-end analysis of two pools of breast cancer and benign breast tissue libraries by SAGE DGED produced 53 differentially expressed genes according to the screening criteria of a >five-fold difference and p<0.01. Further analysis by EST vNorthern and virtual microarray analysis reduced the candidate genes to six, with four down-regulated genes, ANXA1, CAV1, KRT5 and MMP7, and two up-regulated genes, ERBB2 and G1P3 in breast cancer. These findings were validated by a real-time RT-PCR analysis in eight paired human breast cancer tissue samples. We conclude that the combined multiple high throughput analyses is an effective data mining strategy in cancer gene identification. This approach may improve the usage of public available genomic data through strategic data mining of high throughput analysis.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genetic Techniques , Oligonucleotide Array Sequence Analysis/methods , Blotting, Northern , Databases, Genetic , Expressed Sequence Tags , Gene Library , Humans , Pilot Projects , Reverse Transcriptase Polymerase Chain Reaction , Software , Time Factors , Tissue Distribution
16.
Biomed Res Int ; 2015: 325715, 2015.
Article in English | MEDLINE | ID: mdl-26351632

ABSTRACT

BACKGROUND: Studies have shown that young breast cancer patients have more advanced disease and worse survival compared to older patients. Our objective was to study disease characteristics and survival in the subset of young women with hormone receptor positive (HR+) and HER2 negative (HER2-) cancer. METHODS: We retrospectively analyzed HR+/HER2- breast cancer patients who underwent surgery at our institution between 2002 and 2010. We compared clinical characteristics, pathology, treatment, and recurrence-free survival between younger (≤40 years) and older (>40 years) patients. RESULTS: Of 669 HR+/HER2- breast cancer cases, 54 (8.1%) patients were 40 years or younger. Younger patients had more luminal B subtype, high grade, poor differentiation, and increased lymphovascular invasion. Younger women were treated more often with mastectomy and adjuvant chemotherapy. Although the unadjusted recurrence-free survival at median 55-month follow-up was lower in younger women, adjusting for stage, there was no significant difference (90.7% versus 89.3%, p = 0.74) between groups. CONCLUSION: Younger patients with HR+/HER2- breast cancer had more advanced disease and more aggressive treatment than older patients. The unfavorable pathologic features suggest a biologically different tumor in young women. After adjusting for these factors, younger patients have a recurrence-free survival similar to older patients.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Adult , Age Factors , Aged , Chemotherapy, Adjuvant/methods , Disease-Free Survival , Female , Humans , Mastectomy/methods , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Retrospective Studies
17.
Int J Oncol ; 20(6): 1305-11, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12012014

ABSTRACT

MUC1 mucin is aberrantly expressed in many epithelial malignancies and is a promising tumor antigen for target-directed immunotherapy against human breast cancer. Mycobacterium BCG is an effective immunoadjuvant which is known to induce Th1 immune response. Recombinant BCG expressing tumor antigen and secreting cytokine may therefore potentiate the tumor antigen-specific immune responses. In this study, we constructed a recombinant BCG-MUC1-IL2, which expresses a high level of human MUC1 VNTR core protein and secretes functional interleukin 2 (IL2). The immune responses induced by BCG-MUC1-IL2 were examined using a SCID mouse model reconstituted with immunologically competent human lymphocytes, SCID/hu-PBL. The mucin-specific IFN-gamma was secreted only by the lymphocytes derived from animals immunized with BCG-MUC1-IL2, but not with BCG-vector or purified mucin protein for the vaccination. In contrast, in vitro secretion of IL4 by the immunized lymphocytes was only seen in the group of animals which received native MUC1 protein, but not BCG-MUC1-IL2 and BCG-vector. Minimal MUC1-specific IgG and IgM were detected in SCID/hu-PBL mice vaccinated with BCG-MUC1-IL2. These results suggest that BCG-MUC1-IL2 preferentially induces MUC1-specific cellular immune responses and it may serve as a vaccine for breast cancer prevention and treatment.


Subject(s)
Cancer Vaccines/immunology , Interleukin-2/metabolism , Mucin-1/immunology , Vaccines, Synthetic/immunology , Animals , Female , Immunity, Cellular , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Mice , Mice, SCID
18.
Int J Proteomics ; 2014: 390781, 2014.
Article in English | MEDLINE | ID: mdl-25478227

ABSTRACT

Because of the absence of a clear therapeutic target for triple negative breast cancer (TNBC), conventional chemotherapy is the only available systemic treatment option for these patients. Despite chemotherapy treatment, TNBC patients still have worse prognosis when compared with other breast cancer patients. The study is to investigate unique phosphorylated proteins expressed in chemoresistant TNBC cell lines. In the current study, twelve TNBC cell lines were subjected to drug sensitivity assays against chemotherapy drugs docetaxel, doxorubicin, gemcitabine, and cisplatin. Based on their half maximal inhibitory concentrations, four resistant and two sensitive cell lines were selected for further analysis. The phosphopeptides from these cells were enriched with TiO2 beads and fractionated using strong cation exchange. 1,645 phosphoprotein groups and 9,585 unique phosphopeptides were identified by a high throughput LC-MS/MS system LTQ-Orbitrap. The phosphopeptides were further filtered with Ascore system and 1,340 phosphoprotein groups, 2,760 unique phosphopeptides, and 4,549 unique phosphosites were identified. Our study suggested that differentially phosphorylated Cdk5, PML, AP-1, and HSF-1 might work together to promote vimentin induced epithelial to mesenchymal transition (EMT) in the drug resistant cells. EGFR and HGF were also shown to be involved in this process.

19.
Breast Cancer (Auckl) ; 5: 175-99, 2011.
Article in English | MEDLINE | ID: mdl-21863131

ABSTRACT

Notorious for its poor prognosis and aggressive nature, triple-negative breast cancer (TNBC) is a heterogeneous disease entity. The nature of its biological specificity, which is similar to basal-like cancers, tumors arising in BRCA1 mutation carriers, and claudin-low cancers, is currently being explored in hopes of finding the targets for novel biologics and chemotherapeutic agents. In this review, we aim to give a broad overview of the disease's nomenclature and epidemiology, as well as the basic mechanisms of emerging targeted therapies and their performance in clinical trials to date.

20.
Int J Proteomics ; 2011: 896476, 2011.
Article in English | MEDLINE | ID: mdl-22110952

ABSTRACT

Protein-based markers that classify tumor subtypes and predict therapeutic response would be clinically useful in guiding patient treatment. We investigated the LC-MS/MS-identified protein biosignatures in 39 baseline breast cancer specimens including 28 HER2-positive and 11 triple-negative (TNBC) tumors. Twenty proteins were found to correctly classify all HER2 positive and 7 of the 11 TNBC tumors. Among them, galectin-3-binding protein and ALDH1A1 were found preferentially elevated in TNBC, whereas CK19, transferrin, transketolase, and thymosin ß4 and ß10 were elevated in HER2-positive cancers. In addition, several proteins such as enolase, vimentin, peroxiredoxin 5, Hsp 70, periostin precursor, RhoA, cathepsin D preproprotein, and annexin 1 were found to be associated with the tumor responses to treatment within each subtype. The MS-based proteomic findings appear promising in guiding tumor classification and predicting response. When sufficiently validated, some of these candidate protein markers could have great potential in improving breast cancer treatment.

SELECTION OF CITATIONS
SEARCH DETAIL