Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Breast Cancer Res ; 21(1): 141, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31829259

ABSTRACT

Organoid cultures are increasingly used to model human cancers experimentally with a view to tailoring personalized medicine and predicting drug responses. Breast cancer is no exception, but in particular, primary breast cancer poses some inherent difficulties due to the frequent presence of residual non-malignant cells in the biopsies. We originally developed an assay for the distinction between malignant and non-malignant structures in primary breast cancer organoid cultures (Petersen et al., Proc Natl Acad Sci (USA) 89(19):9064-8, 1992). Here, we apply this assay to assess the frequency of normal-like organoids in primary breast carcinoma cultures and the cellular composition as a consequence of passaging. We find that in consecutively collected samples of primary human breast cancers, residual non-malignant tissues were observed histologically in five out of ten biopsies. Based on relevant morphogenesis and correct polarization as recorded by expression in luminal epithelial cells of mucin 1 (Muc1), occludin, and keratin 19 (K19) and expression in basal cells of integrin ß4, p63, and K14, non-malignant organoids were present in all primary human breast cancer-derived cultures. Furthermore, passaging in a contemporary culture medium was in favor of the selective expansion of basal-like cells. We conclude that organoid cultures of human breast cancers are most representative of the tissue origin in primary culture.


Subject(s)
Breast Neoplasms/pathology , Organoids/pathology , Biomarkers , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Fluorescent Antibody Technique , Genetic Variation , Humans , Immunohistochemistry , Tissue Culture Techniques , Tumor Cells, Cultured , Whole Genome Sequencing
2.
Mol Cell Proteomics ; 13(7): 1814-27, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24742827

ABSTRACT

Ferritin heavy chain (FTH1) is a 21-kDa subunit of the ferritin complex, known for its role in iron metabolism, and which has recently been identified as a favorable prognostic protein for triple negative breast cancer (TNBC) patients. Currently, it is not well understood how FTH1 contributes to an anti-tumor response. Here, we explored whether expression and cellular compartmentalization of FTH1 correlates to an effective immune response in TNBC patients. Analysis of the tumor tissue transcriptome, complemented with in silico pathway analysis, revealed that FTH1 was an integral part of an immunomodulatory network of cytokine signaling, adaptive immunity, and cell death. These findings were confirmed using mass spectrometry (MS)-derived proteomic data, and immunohistochemical staining of tissue microarrays. We observed that FTH1 is localized in both the cytoplasm and/or nucleus of cancer cells. However, high cytoplasmic (c) FTH1 was associated with favorable prognosis (Log-rank p = 0.001), whereas nuclear (n) FTH1 staining was associated with adverse prognosis (Log-rank p = 0.019). cFTH1 staining significantly correlated with total FTH1 expression in TNBC tissue samples, as measured by MS analysis (Rs = 0.473, p = 0.0007), but nFTH1 staining did not (Rs = 0.197, p = 0.1801). Notably, IFN γ-producing CD8+ effector T cells, but not CD4+ T cells, were preferentially enriched in tumors with high expression of cFTH1 (p = 0.02). Collectively, our data provide evidence toward new immune regulatory properties of FTH1 in TNBC, which may facilitate development of novel therapeutic targets.


Subject(s)
Apoferritins/metabolism , Biomarkers, Tumor/metabolism , CD8-Positive T-Lymphocytes/immunology , Ferritins/metabolism , Triple Negative Breast Neoplasms/metabolism , Adult , Aged , Apoferritins/biosynthesis , Apoferritins/immunology , Cell Nucleus/metabolism , Cytoplasm/metabolism , Female , Ferritins/biosynthesis , Ferritins/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Middle Aged , Oxidoreductases , Prognosis , Protein Interaction Maps , Proteomics , Tissue Array Analysis , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/mortality
3.
Am J Pathol ; 184(4): 1198-208, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24655379

ABSTRACT

Elucidating the phenotypic evolution of breast cancer through distinct subtypes relies heavily on defining a lineage blueprint of the normal human breast. Here, we show that in normal breast, within the luminal epithelial lineage, a subset of cells characterized by strong staining for endocrine receptors are also characterized by expression of the surface marker CEACAM6. Topographically, this pattern of staining predominates in terminal ductal lobular units, rather than in interlobular ducts. In culture, CEACAM6-expressing cells remain essentially postmitotic under conditions in which the other cells of luminal epithelial lineage are highly proliferative. We examined the pattern of expression among three major breast cancer subtypes: luminal, HER2-enriched, and basal-like. In 104 biopsies, the luminal and HER2-enriched subtypes showed a high proportion of CEACAM6(+) tumors (78% and 83%, respectively); the basal-like subtype showed a low proportion (28%). Further accentuation of this pattern was observed in 13 established breast cancer cell lines. When differentiation was induced by all-trans retinoic acid, CEACAM6 expression strongly correlated with luminal-like differentiation. Furthermore, CEACAM6(+) cancer cells were less proliferative than CEACAM6(-) cells in tumorsphere assays and were less tumorigenic in nude mice. Based on these observations, we propose that luminal and HER2-enriched breast cancers are more closely related than previously thought and may share a common cell of origin.


Subject(s)
Antigens, CD/biosynthesis , Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion Molecules/biosynthesis , Receptor, ErbB-2/metabolism , Animals , Blotting, Western , Female , Flow Cytometry , GPI-Linked Proteins/biosynthesis , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Nude , Real-Time Polymerase Chain Reaction
4.
BMC Cancer ; 15: 35, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25655188

ABSTRACT

BACKGROUND: Human epidermal growth factor receptor-2 (HER2) overexpression and gene amplification are currently established by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH), respectively. This study investigates whether high-density single nucleotide polymorphism (SNP) arrays can provide additional diagnostic power to assess HER2 gene status. METHODS: DNA from 65 breast tumor samples previously diagnosed by HER2 IHC and FISH analysis were blinded and examined for HER2 copy number variation employing SNP array analysis. RESULTS: SNP array analysis identified 24 (37%) samples with selective amplification or imbalance of the HER2 region in the q-arm of chromosome 17. In contrast, only 15 (23%) tumors were found to have HER2 amplification by IHC and FISH analysis. In total, there was a discrepancy in 19 (29%) samples between SNP array and IHC/FISH analysis. In 12 of these cases, the discrepancy towards FISH could be attributed to concomitant amplification or deletion of the centromeric region, which harbors the FISH reference probe sequence. In 3 tumors, repeated IHC/FISH analysis revealed that the original IHC/FISH analysis had failed to indicate the correct HER2 expression level. Finally, the SNP array analysis revealed that more than two thirds of the samples exhibited polyploidy that was unrecognized by conventional FISH. CONCLUSIONS: Collectively, the data show that determination of HER2 copy number variations by SNP array-based genomic segmentation analysis is an effective supplement to IHC/FISH HER2 analysis that, by providing additional diagnostic sensitivity and accuracy, may elect more women for targeted treatment with HER2 inhibitors.


Subject(s)
Breast Neoplasms/genetics , Gene Amplification , Genomics , Polymorphism, Single Nucleotide , Polyploidy , Receptor, ErbB-2/genetics , Breast Neoplasms/diagnosis , DNA Copy Number Variations , Female , Genomics/methods , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Receptor, ErbB-2/metabolism
5.
Mol Cell Proteomics ; 12(2): 381-94, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23172894

ABSTRACT

Breast cancer is a very heterogeneous disease, encompassing several intrinsic subtypes with various morphological and molecular features, natural history and response to therapy. Currently, molecular targeted therapies are available for estrogen receptor (ER)(-) and human epidermal growth factor receptor 2 (Her2)-positive breast tumors. However, a significant proportion of primary breast cancers are negative for ER, progesterone receptor (PgR), and Her2, comprising the triple negative breast cancer (TNBC) group. Women with TNBC have a poor prognosis because of the aggressive nature of these tumors and current lack of suitable targeted therapies. As a consequence, the identification of novel relevant protein targets for this group of patients is of great importance. Using a systematic two dimensional (2D) gel-based proteomic profiling strategy, applied to the analysis of fresh TNBC tissue biopsies, in combination with a three-tier orthogonal technology (two dimensional PAGE/silver staining coupled with MS, two dimensional Western blotting, and immunohistochemistry) approach, we aimed to identify targetable protein markers that were present in a significant fraction of samples and that could define therapy-amenable sub-groups of TNBCs. We present here our results, including a large cumulative database of proteins based on the analysis of 78 TNBCs, and the identification and validation of one specific protein, Mage-A4, which was expressed in a significant fraction of TNBC and Her2-positive/ER negative lesions. The high level expression of Mage-A4 in the tumors studied allowed the detection of the protein in the tumor interstitial fluids as well as in sera. The existence of immunotherapeutics approaches specifically targeting this protein, or Mage-A protein family members, and the fact that we were able to detect its presence in serum suggest novel management options for TNBC and human epidermal growth factor receptor 2 positive/estrogen receptor negative patients bearing Mage-A4 positive tumors.


Subject(s)
Antigens, Neoplasm/blood , Biomarkers, Tumor/blood , Breast Neoplasms/genetics , Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/blood , Proteome/metabolism , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Carcinoma/diagnosis , Carcinoma/metabolism , Electrophoresis, Gel, Two-Dimensional , Female , Gene Expression Profiling , Humans , Mass Spectrometry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proteome/chemistry , Proteome/genetics , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Receptors, Progesterone/deficiency , Receptors, Progesterone/genetics
6.
Biochim Biophys Acta ; 1834(11): 2259-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23416532

ABSTRACT

Tumor interstitial fluid (TIF) is a proximal fluid that, in addition to the set of blood soluble phase-borne proteins, holds a subset of aberrantly externalized components, mainly proteins, released by tumor cells and tumor microenvironment through various mechanisms, which include classical secretion, non-classical secretion, secretion via exosomes and membrane protein shedding. Consequently, the interstitial aqueous phase of solid tumors is a highly promising resource for the discovery of molecules associated with pathological changes in tissues. Firstly, it allows one to delve deeper into the regulatory mechanisms and functions of secretion-related processes in tumor development. Secondly, the anomalous secretion of molecules that is innate to tumors and the tumor microenvironment, being associated with cancer progression, offers a valuable source for biomarker discovery and possible targets for therapeutic intervention. Here we provide an overview of the features of tumor-associated interstitial fluids, based on recent and updated information obtained mainly from our studies of breast cancer. Data from the study of interstitial fluids recovered from several other types of cancer are also discussed. This article is a part of a Special Issue entitled: The Updated Secretome.


Subject(s)
Biomarkers, Tumor/metabolism , Extracellular Fluid/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Proteome/metabolism , Proteomics/methods , Animals , Biomarkers, Tumor/analysis , Humans , Proteome/analysis
7.
Am J Pathol ; 176(3): 1229-40, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20133812

ABSTRACT

Tumor cells can activate stroma, yet the implication of this activation in terms of reciprocal induction of gene expression in tumor cells is poorly understood. Epithelial Stromal Interaction 1 (EPSTI1) is an interferon response gene originally isolated from heterotypic recombinant cultures of human breast cancer cells and activated breast myofibroblasts. Here we describe the first immunolocalization of EPSTI1 in normal and cancerous breast tissue, and we provide evidence for a role of this molecule in the regulation of tumor cell properties and epithelial-mesenchymal transition. In general, no EPSTI1 staining was observed in normal breast epithelial cells from reduction mammoplasties (n=25). However, in carcinomas, staining was positive in 22 of 40 biopsies and inversely correlated with the level of differentiation. To address the function of EPSTI1, we expressed EPSTI1 ectopically in one cell line and silenced endogenous EPSTI1 by RNA interference in another. Irrespective of the experimental approach, EPSTI1 expression led to an increase in tumorsphere formation-a property associated with breast stem/progenitor cells. Most remarkably, we show that EPSTI1, by conveying spread of tumor cells, can replace peritumoral activated fibroblasts in a tumor environment assay. These observations implicate EPSTI1 as a hitherto unappreciated regulator of tumor cell properties.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Neoplasm Proteins/metabolism , Animals , Biological Assay , Cell Line, Tumor , Cell Nucleus/metabolism , Epithelium/metabolism , Epithelium/pathology , Female , Humans , Mesoderm/metabolism , Mesoderm/pathology , Mice , Organ Specificity , RNA Interference
8.
J Proteome Res ; 9(8): 3941-53, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20527979

ABSTRACT

Breast cancer is the leading cause of cancer deaths in women today and is the most common cancer (excluding skin cancers) among women in the Western world. Although cancers detected by screening mammography are significantly smaller than nonscreening ones, noninvasive biomarkers for detection of breast cancer as early as possible are an urgent need as the risk of recurrence and subsequent death is closely related to the stage of the disease at the time of primary surgery. A set of 123 primary breast tumors and matched normal tissue was analyzed by two-dimensional (2D) gel electrophoresis, and a novel protein, C7orf24, was identified as being upregulated in cancer cells. Protein expression levels of C7orf24 were evaluated by immunohistochemical assays to qualify deregulation of this protein. Analysis of C7orf24 expression showed up-regulation in 36.4 and 23.4% of cases present in the discovery sample set (123 samples) and in an independent large TMA validation data set (2197 samples) of clinically annotated breast cancer specimens, respectively. Survival analysis showed that C7orf24 overexpression defines a subgroup of breast tumors with poor clinical outcome. Up-regulation of C7orf24 was also found in other cancer types. Four of these were investigated in greater detail, and we found that a proportion of tumors (58% in cervical, 38% in lung, 72% in colon, and 46% in breast cancer) expressed C7orf24 at levels exceeding those seen in normal samples. The observed overexpression of this protein in different types of cancer suggests deregulation of C7orf24 to be a general event in epithelial carcinogenesis, indicating that this protein may play an important role in cancer cell biology and thus constitute a novel therapeutic target. Furthermore, as C7orf24 is externalized to the tissue extracellular fluid and can be detected in serum, this protein also represents a potential serological marker.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/diagnosis , Carcinoma/diagnosis , Gene Expression Regulation, Neoplastic/genetics , gamma-Glutamylcyclotransferase/metabolism , Biomarkers, Tumor/blood , Breast Neoplasms/metabolism , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Survival Analysis , Uterine Cervical Neoplasms/metabolism , gamma-Glutamylcyclotransferase/genetics
9.
Mol Cell Proteomics ; 7(10): 1795-809, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18632593

ABSTRACT

Established histopathological criteria divide invasive breast carcinomas into defined groups. Ductal of no specific type and lobular are the two major subtypes accounting for around 75 and 15% of all cases, respectively. The remaining 10% include rarer types such as tubular, cribriform, mucinous, papillary, medullary, metaplastic, and apocrine breast carcinomas. Molecular profiling technologies, on the other hand, subdivide breast tumors into five subtypes, basal-like, luminal A, luminal B, normal breast tissue-like, and ERBB2-positive, that have different prognostic characteristics. An additional subclass termed "molecular apocrine" has recently been described, but these lesions did not exhibit all the histopathological features of classical invasive apocrine carcinomas (IACs). IACs make up 0.5-3% of the invasive ductal carcinomas, and despite the fact that they are morphologically distinct from other breast lesions, there are presently no standard molecular criteria available for their diagnosis and as a result no precise information as to their prognosis. Toward this goal our laboratories have embarked in a systematic proteomics endeavor aimed at identifying biomarkers that may characterize and subtype these lesions as well as targets that may lead to the development of novel targeted therapies and chemoprevention strategies. By comparing the protein expression profiles of apocrine macrocysts and non-malignant breast epithelial tissue we have previously reported the identification of a few proteins that are specifically expressed by benign apocrine lesions as well as by the few IACs that were available to us at the time. Here we reiterate our strategy to reveal apocrine cell markers and present novel data, based on the analysis of a considerably larger number of samples, establishing that IACs correspond to a distinct molecular subtype of breast carcinomas characterized by the expression of 15-prostaglandin dehydrogenase alone or in combination with a novel form of acyl-CoA synthetase medium-chain family member 1 (ACSM1). Moreover we show that 15-prostaglandin dehydrogenase is not expressed by other breast cancer types as determined by gel-based proteomics and immunohistochemistry analysis and that antibodies against this protein can identify IACs in an unbiased manner in a large breast cancer tissue microarray making them potentially useful as a diagnostic aid.


Subject(s)
Apocrine Glands/enzymology , Apocrine Glands/pathology , Breast Neoplasms/classification , Breast Neoplasms/enzymology , Coenzyme A Ligases/metabolism , Hydroxyprostaglandin Dehydrogenases/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Cohort Studies , Disease Progression , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunohistochemistry , Immunophenotyping , Middle Aged , Neoplasm Invasiveness , Paraffin Embedding , Phenotype , Silver Staining , Tissue Array Analysis
10.
Sci Rep ; 10(1): 4788, 2020 Mar 11.
Article in English | MEDLINE | ID: mdl-32161318

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Breast Cancer Res Treat ; 115(1): 69-76, 2009 May.
Article in English | MEDLINE | ID: mdl-18500671

ABSTRACT

Germ-line mutations in the tumour suppressor proteins BRCA1 and BRCA2 predispose to breast and ovarian cancer. We examined 32 breast and/or ovarian cancer patients from Greenland for mutations in BRCA1 and BRCA2. Whereas no mutations were identified in 19 families, 13 families exhibited a BRCA1 exon 3 nucleotide 234 T > G mutation, which has not previously been reported in the breast cancer information core (BIC) database. The mutation changes a conserved cysteine 39 to a glycine in the Zn(2+) site II of the RING domain, which is essential for BRCA1 ubiquitin ligase activity. Eight of the families had members with ovarian cancer, suggesting that the RING domain may be an ovarian cancer hotspot. By SNP array analysis, we find that all 13 families share a 4.5 Mb genomic fragment containing the BRCA1 gene, showing that the mutation originates from a founder. Finally, analysis of 1152 Inuit, representing almost ~2% of the total Greenlandic Inuit population, showed that the frequency of the mutation was 1.0%. We conclude that the BRCA1 nucleotide 234 T > G is a common Greenlandic Inuit founder mutation. The relative high frequency in the general population, together with the ease of screening and possibility to reduce mortality in gene carriers, may warrant screening of the Greenlandic Inuit population. Provided screening is efficient, about 5% of breast- and 13% of ovarian cancers, respectively, may be prevented.


Subject(s)
Founder Effect , Genes, BRCA1 , Mutation , Adult , Amino Acid Sequence , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Female , Genes, BRCA2 , Greenland , Humans , Inuit , Middle Aged , Molecular Sequence Data , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Polymorphism, Single Nucleotide , Protein Structure, Tertiary , Sequence Homology, Amino Acid
12.
Sci Rep ; 9(1): 14843, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31619692

ABSTRACT

Human breast cancer is believed to arise in luminal progenitors within the normal breast. A subset of these are double positive (DP) for basal and luminal keratins and localizes to a putative stem cell zone within ducts. We here present a new protocol based on a combination of CD146 with CD117 and CD326 which provides an up to thirty fold enrichment of the DP cells. We show by expression profiling, colony formation, and morphogenesis that CD146high/CD117high/CD326high DP cells belong to a luminal progenitor compartment. While these DP cells are located quite uniformly in ducts, with age a variant type of DP (vDP) cells, which is mainly CD146-negative, accumulates in lobules. Intriguingly, in specimens with BRCA1 mutations known to predispose for cancer, higher frequencies of lobular vDP cells are observed. We propose that vDP cells are strong candidates for tracing the cellular origin of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinogenesis , Keratin-14/metabolism , Keratin-19/metabolism , Mammary Glands, Human/metabolism , Neoplastic Stem Cells/metabolism , Adolescent , Adult , Breast Neoplasms/pathology , CD146 Antigen/metabolism , Cells, Cultured , Female , Healthy Volunteers , Humans , Mammary Glands, Human/pathology , Middle Aged , Neoplastic Stem Cells/pathology , Young Adult
13.
Proteomics ; 8(23-24): 5038-52, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19003862

ABSTRACT

Axillary lymph node (ALN) status is currently used as an important clinical indicator of breast cancer prognosis. However, the molecular mechanisms underlying lymph node metastasis are poorly understood and the relationship between ALN metastasis and the primary tumor remains unclear. In an effort to reveal structural changes in the genome and related protein responses that may drive regional metastatic progression we have analyzed matched pairs of primary breast tumors and ALN metastases both at the genomic and proteomic levels using comparative genomic hybridization (CGH) array, quantitative high-resolution 2-D PAGE in combination with MS, and immunohistochemistry (IHC). Array CGH revealed a remarkable similarity in genomic aberration profiles between the matched primary tumors and the ALN metastases. Quantitative profiling of 135 known proteins also revealed striking similarities in their overall expression patterns, although we observed distinct changes in the levels of individual proteins in some sample pairs. The remarkable similarities of the overall genomic and proteomic profiles between primary tumors and matched ALN metastases are taken to suggest that, in general, key biological characteristics of the primary breast tumor are maintained in the corresponding lymph node metastases. Given that the omics-based technologies are oblivious to changes that only occur in minor cellular subsets we validated the proteomic data using IHC, which provides protein expression information with a valuable topological component. Besides confirming the omics-derived data, the IHC analysis revealed that in two cases the ALN metastases may have been derived from a distinct minor cell subpopulation present in the primary tumor rather than from the bulk of it.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Profiling , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Proteomics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Comparative Genomic Hybridization , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunohistochemistry , Male , Middle Aged , Models, Biological , Neoplasm Proteins/analysis , Paraffin Embedding
14.
Oncoimmunology ; 5(12): e1248015, 2016.
Article in English | MEDLINE | ID: mdl-28123884

ABSTRACT

The tumor microenvironment is composed of many immune cell subpopulations and is an important factor in the malignant progression of neoplasms, particularly breast cancer (BC). However, the cytokine networks that coordinate various regulatory events within the BC interstitium remain largely uncharacterized. Moreover, the data obtained regarding the origin of cytokine secretions, the levels of secretion associated with tumor development, and the possible clinical relevance of cytokines remain controversial. Therefore, we profiled 27 cytokines in 78 breast tumor interstitial fluid (TIF) samples, 43 normal interstitial fluid (NIF) samples, and 25 matched serum samples obtained from BC patients with Luminex xMAP multiplex technology. Eleven cytokines exhibited significantly higher levels in the TIF samples compared with the NIF samples: interleukin (IL)-7, IL-10, fibroblast growth factor-2, IL-13, interferon (IFN)γ-inducible protein (IP-10), IL-1 receptor antagonist (IL-1RA), platelet-derived growth factor (PDGF)-ß, IL-1ß, chemokine ligand 5 (RANTES), vascular endothelial growth factor, and IL-12. An immunohistochemical analysis further demonstrated that IL-1RA, IP-10, IL-10, PDGF-ß, RANTES, and VEGF are widely expressed by both cancer cells and tumor-infiltrating lymphocytes (TILs), whereas IP-10 and RANTES were preferentially abundant in triple-negative breast cancers (TNBCs) compared to Luminal A subtype cancers. The latter observation corresponds with the high level of TILs in the TNBC samples. IL-1ß, IL-7, IL-10, and PDGFß also exhibited a correlation between the TIF samples and matched sera. In a survival analysis, high levels of IL-5, a hallmark TH2 cytokine, in the TIF samples were associated with a worse prognosis. These findings have important implications for BC immunotherapy research.

15.
Sci Rep ; 6: 32027, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27558661

ABSTRACT

We have previously identified UMP-CMP kinase (CMPK1) as a prognostic marker for triple negative breast cancer (TNBC) by mass spectrometry (MS). In this study we evaluated CMPK1 association to prognosis in an independent set of samples by immunohistochemistry (IHC) and assessed biological pathways associated to its expression through gene set enrichment analysis (GSEA). A total of 461 TNBC paraffin-embedded tissues were collected from different academic hospitals in Europe, incorporated into tissue micro-arrays (TMA), and stained for CMPK1 expression. We also collected gene expression data of 60 samples, which were also present in the TMA, for GSEA correlation analysis. CMPK1 IHC staining showed both cytoplasmic and nuclear components. While cytoplasmic CMPK1 did not show any association to metastasis free survival (MFS), nuclear CMPK1 was associated to poor prognosis independently from other prognostic factors in stratified Cox regression analyses. GSEA correlation analysis of the nuclear CMPK1-stratified gene expression dataset showed a significant enrichment of extracellular matrix (ECM; positive correlation) and cell cycle (negative correlation) associated genes. We have shown here that nuclear CMPK1 is indicative of poor prognosis in TNBCs and that its expression may be related to dysregulation of ECM and cell cycle molecules.


Subject(s)
Nucleoside-Phosphate Kinase/metabolism , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/mortality , Adult , Aged , Cell Nucleus/enzymology , Databases, Factual , Extracellular Matrix/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Nucleoside-Phosphate Kinase/genetics , Prognosis , Tissue Array Analysis , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy
16.
PLoS One ; 9(11): e112024, 2014.
Article in English | MEDLINE | ID: mdl-25389781

ABSTRACT

Apocrine carcinoma of the breast is a distinctive malignancy with unique morphological and molecular features, generally characterized by being negative for estrogen and progesterone receptors, and thus not electable for endocrine therapy. Despite the fact that they are morphologically distinct from other breast lesions, no standard molecular criteria are currently available for their diagnosis. Using gel-based proteomics in combination with mass spectrometry and immunohistochemistry we have identified two novel markers, HMGCS2 and FABP7 that categorize the entire breast apocrine differentiation spectrum from benign metaplasia and cysts to invasive stages. Expression of HMGCS2 and FABP7 is strongly associated with apocrine differentiation; their expression is retained by most invasive apocrine carcinomas (IAC) showing positive immunoreactivity in 100% and 78% of apocrine carcinomas, respectively, as compared to non-apocrine tumors (16.7% and 6.8%). The nuclear localization of FABP7 in tumor cells was shown to be associated with more aggressive stages of apocrine carcinomas. In addition, when added to the panel of apocrine biomarkers previously reported by our group: 15-PGDH, HMGCR and ACSM1, together they provide a signature that may represent a golden molecular standard for defining the apocrine phenotype in the breast. Moreover, we show that combining HMGCS2 to the steroidal profile (HMGCS2+/Androgen Receptor (AR)+/Estrogen Receptor(ER)-/Progesteron Receptor (PR)- identifies IACs with a greater sensitivity (79%) as compared with the steroidal profile (AR+/ER-/PR-) alone (54%). We have also presented a detailed immunohistochemical analysis of breast apocrine lesions with a panel of antibodies against proteins which correspond to 10 genes selected from published transcriptomic signatures that currently characterize molecular apocrine subtype and shown that except for melanophilin that is overexpressed in benign apocrine lesions, these proteins were not specific for morphological apocrine differentiation in breast.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Carcinoma/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Hydroxymethylglutaryl-CoA Synthase/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Antibodies/chemistry , Cell Differentiation , Cell Line, Tumor , Disease Progression , Fatty Acid-Binding Protein 7 , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Middle Aged , Reproducibility of Results
17.
Cardiovasc Pathol ; 20(1): 63-7, 2011.
Article in English | MEDLINE | ID: mdl-20060324

ABSTRACT

INTRODUCTION: We report the clinical, pathological, and immunohistochemical features of four primary malignant cardiac tumors identified at the Department of Pathology, Rigshospitalet, Denmark. A panel of immunohistochemical markers for classification is proposed. METHODS: Between 2000 and 2008, four patients with malignant cardiac tumors were treated at our hospital. We retrospectively reviewed the medical records and evaluated the patient characteristics and treatment. RESULTS: Three patients presented with severe dyspnea; one patient presented with chest pain. Transthoracic echocardiography demonstrated, in all four cases, abnormal masses in the atria. The cases were, based on morphological features and immunoprofile, classified as myogenic sarcoma (two cases), undifferentiated pleomorphic sarcoma, and leiomyosarcoma. Three of the patients received orthotopic heart transplantation. One patient survived 6.5 years after the diagnosis, and two patients are still alive 2 and 3 years after being diagnosed, respectively. CONCLUSIONS: All four cases were sarcomas. A limited number of immunohistochemical markers can be used in order to define a specific line of differentiation. In this small study, three of the patients were offered orthotopic heart transplantation, and the survival times were generally longer than in most series.


Subject(s)
Heart Neoplasms/pathology , Sarcoma/pathology , Adult , Biomarkers/metabolism , Female , Heart Neoplasms/metabolism , Heart Neoplasms/surgery , Heart Transplantation , Humans , Immunohistochemistry , Leiomyosarcoma/metabolism , Leiomyosarcoma/pathology , Leiomyosarcoma/surgery , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Retrospective Studies , Sarcoma/metabolism , Sarcoma/secondary , Sarcoma/surgery
19.
Mol Oncol ; 4(1): 65-89, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20005186

ABSTRACT

Breast cancer is by far the most common diagnosed form of cancer and the leading cause of cancer death in women today. Clinically useful biomarkers for early detection of breast cancer could lead to a significant reduction in mortality. Here we describe a detailed analysis using gel-based proteomics in combination with mass spectrometry and immunohistochemistry (IHC) of the tumour interstitial fluids (TIF) and normal interstitial fluids (NIF) collected from 69 prospective breast cancer patients. The goal of this study was to identify abundant cancer up-regulated proteins that are externalised by cells in the tumour microenvironment of most if not all these lesions. To this end, we applied a phased biomarker discovery research strategy to the analysis of these samples rather than comparing all samples among each other, with inherent inter and intra-sample variability problems. To this end, we chose to use samples derived from a single tumour/benign tissue pair (patient 46, triple negative tumour), for which we had well-matched samples in terms of epithelial cell numbers, to generate the initial dataset. In this first phase we found 110 proteins that were up-regulated by a factor of 2 or more in the TIF, some of which were confirmed by IHC. In the second phase, we carried out a systematic computer assisted analysis of the 2D gels of the remaining 68 TIF samples in order to identify TIF 46 up-regulated proteins that were deregulated in 90% or more of all the available TIFs, thus representing common breast cancer markers. This second phase singled out a set of 26 breast cancer markers, most of which were also identified by a complementary analysis using LC-MS/MS. The expression of calreticulin, cellular retinoic acid-binding protein II, chloride intracellular channel protein 1, EF-1-beta, galectin 1, peroxiredoxin-2, platelet-derived endothelial cell growth factor, protein disulfide isomerase and ubiquitin carboxyl-terminal hydrolase 5 were further validated using a tissue microarray containing 70 malignant breast carcinomas of various grades of atypia. A significant number of these proteins have already been detected in the blood/plasma/secretome by others. The next steps, which include biomarker prioritization based on the hierarchal evaluation of these markers, antibody and antigen development, assay development, analytical validation, and preliminary testing in the blood of healthy and breast cancer patients, are discussed.


Subject(s)
Biomarkers, Tumor/metabolism , Biomarkers/metabolism , Breast Neoplasms/diagnosis , Neoplasm Staging/classification , Body Fluids/chemistry , Breast Neoplasms/metabolism , Calreticulin/metabolism , Chloride Channels/metabolism , Early Diagnosis , Electrophoresis, Gel, Two-Dimensional/methods , Female , Galectin 1/metabolism , Humans , Immunohistochemistry , Neoplasm Staging/standards , Peptide Elongation Factor 1/metabolism , Peroxiredoxins/metabolism , Prognosis , Protein Disulfide-Isomerases/metabolism , Proteomics/statistics & numerical data , Thymidine Phosphorylase/metabolism , Up-Regulation
20.
Mol Oncol ; 4(6): 539-61, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21036680

ABSTRACT

Our limited understanding of the biological impact of the whole spectrum of early breast lesions together with a lack of accurate molecular-based risk criteria for the diagnosis and assignment of prognostic significance to biopsy findings presents an important problem in the clinical management of patients harboring precancerous breast lesions. As a result, there is a need to identify biomarkers that can better determine the outcome of early breast lesions by identifying subpopulations of cells in breast premalignant disease that are at high-risk of progression to invasive disease. A first step towards achieving this goal will be to define the molecular phenotypes of the various cell types and precursors - generated by the stem cell hierarchy - that are present in normal and benign conditions of the breast. To date there have been very few systematic proteomic studies aimed at characterizing the phenotypes of the different cell subpopulations present in normal human mammary tissue, partly due to the formidable heterogeneity of mammary tissue, but also due to limitations of the current proteomic technologies. Work in our laboratories has attempted to address in a systematic fashion some of these limitations and here we present our efforts to search for biomarkers using normal fresh tissue from non-neoplastic breast samples. From the data generated by the 2D gel-based proteomic profiling we were able to compile a protein database of normal human breast epithelial tissue that was used to support the biomarker discovery program. We review and present new data on the putative cell-progenitor marker cytokeratin 15 (CK15), and describe a novel marker, dihydropyriminidase-related protein 3 (DRP3) that in combination with CK15 and other well known proteins were used to define molecular phenotypes of normal human breast epithelial cells and their progenitors in resting acini, lactating alveoli, and large collecting ducts of the nipple. Preliminary results are also presented concerning DRP3 positive usual ductal hyperplasias (UDHs) and on single cell layer columnar cells (CCCs). At least two bona fide biomarkers of undifferentiated ERα/PgR negative luminal cells emerged from these studies, CK15 and c-KIT, which in combination with transformation markers may lead to the establishment of a protein signature able to identify breast precancerous at risk of progressing to invasive disease.


Subject(s)
Biomarkers/metabolism , Breast Neoplasms , Epithelial Cells/metabolism , Mammary Glands, Human , Phenotype , Proteomics/methods , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Databases, Protein , Electrophoresis, Gel, Two-Dimensional/methods , Female , Humans , Immunohistochemistry , Immunophenotyping , Keratin-15/metabolism , Keratin-19/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mass Spectrometry/methods , Muscle Proteins/metabolism , Protein Array Analysis/methods , Protein Isoforms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL