Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Pathol ; 247(3): 287-292, 2019 03.
Article in English | MEDLINE | ID: mdl-30430577

ABSTRACT

Estrogen receptor α-positive (ER-positive) or 'luminal' breast cancers were notoriously difficult to establish as patient-derived xenografts (PDXs). We and others recently demonstrated that the microenvironment is critical for ER-positive tumor cells; when grafted as single cells into milk ducts of NOD Scid gamma females, >90% of ER-positive tumors can be established as xenografts and recapitulate many features of the human disease in vivo. This intraductal approach holds promise for personalized medicine, yet human and murine stroma are organized differently and this and other species specificities may limit the value of this model. Here, we analyzed 21 ER-positive intraductal PDXs histopathologically. We found that intraductal PDXs vary in extent and define four histopathological patterns: flat, lobular, in situ and invasive, which occur in pure and combined forms. The intraductal PDXs replicate earlier stages of tumor development than their clinical counterparts. Micrometastases are already detected when lesions appear in situ. Tumor extent, histopathological patterns and micrometastatic load correlate with biological properties of their tumors of origin. Our findings add evidence to the validity of the intraductal model for in vivo studies of ER-positive breast cancer and raise the intriguing possibility that tumor cell dissemination may occur earlier than currently thought. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Carcinoma, Intraductal, Noninfiltrating/pathology , Estrogen Receptor alpha/metabolism , Mammary Neoplasms, Experimental/pathology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/secondary , Female , Heterografts , Humans , Mammary Neoplasms, Experimental/metabolism , Mice, SCID , Neoplasm Micrometastasis/pathology , Neoplasm Transplantation
2.
Lab Invest ; 97(1): 93-103, 2017 01.
Article in English | MEDLINE | ID: mdl-27892928

ABSTRACT

Fluorescence in situ hybridization (FISH) is one of the recommended techniques for human epidermal growth factor receptor 2 (HER2) status assessment on cancer tissues. Here we develop microfluidics-assisted FISH (MA-FISH), in which hybridization of the FISH probes with their target DNA strands is obtained by applying square-wave oscillatory flows of diluted probe solutions in a thin microfluidic chamber of 5 µl volume. By optimizing the experimental parameters, MA-FISH decreases the consumption of the expensive probe solution by a factor 5 with respect to the standard technique, and reduces the hybridization time to 4 h, which is four times faster than in the standard protocol. To validate the method, we blindly conducted HER2 MA-FISH on 51 formalin-fixed paraffin-embedded tissue slides of 17 breast cancer samples, and compared the results with standard HER2 FISH testing. HER2 status classification was determined according to published guidelines, based on average number of HER2 copies per cell and average HER2/CEP17 ratio. Excellent agreement was observed between the two methods, supporting the validity of MA-FISH and further promoting its short hybridization time and reduced reagent consumption.


Subject(s)
Breast Neoplasms/genetics , In Situ Hybridization, Fluorescence/methods , Microfluidics/methods , Receptor, ErbB-2/genetics , Breast Neoplasms/diagnosis , DNA Probes/genetics , Female , Gene Dosage , Humans , Reproducibility of Results , Sensitivity and Specificity , Time Factors
3.
Biochim Biophys Acta ; 1836(1): 146-57, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23628726

ABSTRACT

HER2 gene amplification is observed in about 15% of breast cancers. The subgroup of HER2-positive breast cancers appears to be heterogeneous and presents complex patterns of gene amplification at the locus on chromosome 17q12-21. The molecular variations within the chromosome 17q amplicon and their clinical implications remain largely unknown. Besides the well-known TOP2A gene encoding Topoisomerase IIA, other genes might also be amplified and could play functional roles in breast cancer development and progression. This review will focus on the current knowledge concerning the HER2 amplicon heterogeneity, its clinical and biological impact and the pitfalls associated with the evaluation of gene amplifications at this locus, with particular attention to TOP2A and the link between TOP2A and anthracycline benefit. In addition it will discuss the clinical and biological implications of the amplification of ten other genes at this locus (MED1, STARD3, GRB7, THRA, RARA, IGFPB4, CCR7, KRT20, KRT19 and GAST) in breast cancer.


Subject(s)
Anthracyclines/therapeutic use , Antigens, Neoplasm/metabolism , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Gene Amplification , Receptor, ErbB-2/genetics , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Female , Humans , Poly-ADP-Ribose Binding Proteins
4.
Ann Pathol ; 34(5): 366-72, 2014 Oct.
Article in French | MEDLINE | ID: mdl-25439989

ABSTRACT

Biomarker assessment of breast cancer tumor samples is part of the routine workflow of pathology laboratories. International guidelines have recently been updated, with special regards to the pre-analytical steps that are critical for the quality of immunohistochemical and in situ hybridization procedures, whatever the biomarker analyzed. Fixation and specimen handling protocols must be standardized, validated and carefully tracked. Cooperation and training of the personnel involved in the specimen workflow (e.g. radiologists, surgeons, nurses, technicians and pathologists) are of paramount importance. The GEFPICS' update of the recommendations herein details and comments the different steps of the pre-analytical process. Application of these guidelines and participation to quality insurance programs are mandatory to ensure the correct evaluation of oncotheranostic biomarkers.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Immunohistochemistry/methods , In Situ Hybridization/methods , Receptor, ErbB-2/analysis , Receptors, Steroid/analysis , Breast Neoplasms/pathology , Female , Fixatives , France , Histological Techniques , Humans , Prognosis , Quality Control , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Specimen Handling/methods
5.
Ann Pathol ; 34(5): 352-65, 2014 Oct.
Article in French | MEDLINE | ID: mdl-25439988

ABSTRACT

International guidelines on HER2 determination in breast cancer have just been updated by the American Society of Clinical Oncology (ASCO) and College of American Pathologists (CAP), on the basis of more than ten-year practice, results of clinical trials and concordance studies. The GEFPICS group, composed of expert pathologists in breast cancer, herein presents these recommendations, adapted to the French routine practice. These guidelines highlight the possible diagnosis difficulties with regards to HER2 status determination, such as intra-tumor heterogeneity, special histological subtypes and biomarker re-evaluation during metastatic relapse. Pre-analytical issues and updated scoring criteria (especially for equivocal cases) are detailed, in order to decrease the occurrence of false negative cases. In the era of personalized medicine, pathologists are more than ever involved in the quality of oncotheranostic biomarker evaluation.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Receptor, ErbB-2/analysis , Breast Neoplasms/drug therapy , False Negative Reactions , Female , France , Humans , Immunohistochemistry/methods , In Situ Hybridization , In Situ Hybridization, Fluorescence , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local , Prognosis
6.
Int J Gynecol Pathol ; 31(3): 236-41, 2012 May.
Article in English | MEDLINE | ID: mdl-22498940

ABSTRACT

In 2004, a 56-year-old woman was diagnosed with Stage IA follicular lymphoma in a cervical lymph node biopsy. The patient experienced total remission after local radiation therapy. In 2009, a control computed tomography scan evidenced a pelvic mass, prompting total hysterectomy. The latter harbored a 4.8-cm intramural uterine tumor corresponding to a mostly diffuse and focally nodular proliferation of medium to large cells, with extensive, periodic acid-Schiff negative, signet ring cell changes, and a pan-keratin negative, CD20+, CD10+, Bcl2+, Bcl6+ immunophenotype. Molecular genetic studies showed the same clonal IGH gene rearrangement in the lymph node and the uterus, establishing the uterine tumor as a relapse of the preceding follicular lymphoma, although no signet ring cells were evidenced at presentation. Uterine localization of lymphomas is rare, and lymphomas with signet ring cell features are uncommon. This exceptional case exemplifies a diagnostically challenging situation and expands the differential diagnosis of uterine neoplasms displaying signet ring cell morphology.


Subject(s)
Carcinoma, Signet Ring Cell/diagnosis , Lymphoma, B-Cell/diagnosis , Uterine Neoplasms/diagnosis , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Signet Ring Cell/pathology , Carcinoma, Signet Ring Cell/therapy , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Follow-Up Studies , Humans , Hysterectomy , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/therapy , Middle Aged , Prednisone/administration & dosage , Treatment Outcome , Uterine Neoplasms/pathology , Uterine Neoplasms/therapy , Vincristine/administration & dosage
7.
Nat Commun ; 13(1): 3127, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35668111

ABSTRACT

Estrogen and progesterone receptor (ER, PR) signaling control breast development and impinge on breast carcinogenesis. ER is an established driver of ER + disease but the role of the PR, itself an ER target gene, is debated. We assess the issue in clinically relevant settings by a genetic approach and inject ER + breast cancer cell lines and patient-derived tumor cells to the milk ducts of immunocompromised mice. Such ER + xenografts were exposed to physiologically relevant levels of 17-ß-estradiol (E2) and progesterone (P4). We find that independently both premenopausal E2 and P4 levels increase tumor growth and combined treatment enhances metastatic spread. The proliferative responses are patient-specific with MYC and androgen receptor (AR) signatures determining P4 response. PR is required for tumor growth in patient samples and sufficient to drive tumor growth and metastasis in ER signaling ablated tumor cells. Our findings suggest that endocrine therapy may need to be personalized, and that abrogating PR expression can be a therapeutic option.


Subject(s)
Breast Neoplasms , Receptors, Progesterone , Animals , Breast Neoplasms/metabolism , Estradiol/pharmacology , Estradiol/therapeutic use , Female , Humans , Mice , Progesterone/pharmacology , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
8.
Med Image Anal ; 75: 102264, 2022 01.
Article in English | MEDLINE | ID: mdl-34781160

ABSTRACT

Cancer diagnosis, prognosis, and therapy response predictions from tissue specimens highly depend on the phenotype and topological distribution of constituting histological entities. Thus, adequate tissue representations for encoding histological entities is imperative for computer aided cancer patient care. To this end, several approaches have leveraged cell-graphs, capturing the cell-microenvironment, to depict the tissue. These allow for utilizing graph theory and machine learning to map the tissue representation to tissue functionality, and quantify their relationship. Though cellular information is crucial, it is incomplete alone to comprehensively characterize complex tissue structure. We herein treat the tissue as a hierarchical composition of multiple types of histological entities from fine to coarse level, capturing multivariate tissue information at multiple levels. We propose a novel multi-level hierarchical entity-graph representation of tissue specimens to model the hierarchical compositions that encode histological entities as well as their intra- and inter-entity level interactions. Subsequently, a hierarchical graph neural network is proposed to operate on the hierarchical entity-graph and map the tissue structure to tissue functionality. Specifically, for input histology images, we utilize well-defined cells and tissue regions to build HierArchical Cell-to-Tissue (HACT) graph representations, and devise HACT-Net, a message passing graph neural network, to classify the HACT representations. As part of this work, we introduce the BReAst Carcinoma Subtyping (BRACS) dataset, a large cohort of Haematoxylin & Eosin stained breast tumor regions-of-interest, to evaluate and benchmark our proposed methodology against pathologists and state-of-the-art computer-aided diagnostic approaches. Through comparative assessment and ablation studies, our proposed method is demonstrated to yield superior classification results compared to alternative methods as well as individual pathologists. The code, data, and models can be accessed at https://github.com/histocartography/hact-net.


Subject(s)
Histological Techniques , Neural Networks, Computer , Benchmarking , Humans , Prognosis
9.
J Pathol Clin Res ; 8(2): 191-205, 2022 03.
Article in English | MEDLINE | ID: mdl-34889530

ABSTRACT

Invasive lobular breast carcinoma (ILC) is the second most common breast carcinoma (BC) subtype and is mainly driven by loss of E-cadherin expression. Correct classification of BC as ILC is important for patient treatment. This study assessed the degree of agreement among pathologists for the diagnosis of ILC. Two sets of hormone receptor (HR)-positive/HER2-negative BCs were independently reviewed by participating pathologists. In set A (61 cases), participants were provided with hematoxylin/eosin (HE)-stained sections. In set B (62 cases), participants were provided with HE-stained sections and E-cadherin immunohistochemistry (IHC). Tumor characteristics were balanced. Participants classified specimens as non-lobular BC versus mixed BC versus ILC. Pairwise inter-observer agreement and agreement with a pre-defined reference diagnosis were determined with Cohen's kappa statistics. Subtype calls were correlated with molecular features, including CDH1/E-cadherin mutation status. Thirty-five pathologists completed both sets, providing 4,305 subtype calls. Pairwise inter-observer agreement was moderate in set A (median κ = 0.58, interquartile range [IQR]: 0.48-0.66) and substantial in set B (median κ = 0.75, IQR: 0.56-0.86, p < 0.001). Agreement with the reference diagnosis was substantial in set A (median κ = 0.67, IQR: 0.57-0.75) and almost perfect in set B (median κ = 0.86, IQR: 0.73-0.93, p < 0.001). The median frequency of CDH1/E-cadherin mutations in specimens classified as ILC was 65% in set A (IQR: 56-72%) and 73% in set B (IQR: 65-75%, p < 0.001). Cases with variable subtype calls included E-cadherin-positive ILCs harboring CDH1 missense mutations, and E-cadherin-negative ILCs with tubular elements and focal P-cadherin expression. ILCs with trabecular growth pattern were often misclassified as non-lobular BC in set A but not in set B. In conclusion, subtyping of BC as ILC achieves almost perfect agreement with a pre-defined reference standard, if assessment is supported by E-cadherin IHC. CDH1 missense mutations associated with preserved E-cadherin protein expression, E- to P-cadherin switching in ILC with tubular elements, and trabecular ILC were identified as potential sources of discordant classification.


Subject(s)
Breast Neoplasms , Carcinoma, Lobular , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Lobular/diagnosis , Carcinoma, Lobular/genetics , Female , Humans , Immunohistochemistry , Observer Variation
10.
EMBO Mol Med ; 13(7): e14314, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34042278

ABSTRACT

Hormonal contraception exposes women to synthetic progesterone receptor (PR) agonists, progestins, and transiently increases breast cancer risk. How progesterone and progestins affect the breast epithelium is poorly understood because we lack adequate models to study this. We hypothesized that individual progestins differentially affect breast epithelial cell proliferation and hence breast cancer risk. Using mouse mammary tissue ex vivo, we show that testosterone-related progestins induce the PR target and mediator of PR signaling-induced cell proliferation receptor activator of NF-κB ligand (Rankl), whereas progestins with anti-androgenic properties in reporter assays do not. We develop intraductal xenografts of human breast epithelial cells from 36 women, show they remain hormone-responsive and that progesterone and the androgenic progestins, desogestrel, gestodene, and levonorgestrel, promote proliferation but the anti-androgenic, chlormadinone, and cyproterone acetate, do not. Prolonged exposure to androgenic progestins elicits hyperproliferation with cytologic changes. Androgen receptor inhibition interferes with PR agonist- and levonorgestrel-induced RANKL expression and reduces levonorgestrel-driven cell proliferation. Thus, different progestins have distinct biological activities in the breast epithelium to be considered for more informed choices in hormonal contraception.


Subject(s)
Androgens , Progestins , Animals , Cell Proliferation , Contraceptive Agents , Mice
11.
EMBO Mol Med ; 13(3): e13180, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33616307

ABSTRACT

Invasive lobular carcinoma (ILC) is the most frequent special histological subtype of breast cancer, typically characterized by loss of E-cadherin. It has clinical features distinct from other estrogen receptor-positive (ER+ ) breast cancers but the molecular mechanisms underlying its characteristic biology are poorly understood because we lack experimental models to study them. Here, we recapitulate the human disease, including its metastatic pattern, by grafting ILC-derived breast cancer cell lines, SUM-44 PE and MDA-MB-134-VI cells, into the mouse milk ducts. Using patient-derived intraductal xenografts from lobular and non-lobular ER+ HER2- tumors to compare global gene expression, we identify extracellular matrix modulation as a lobular carcinoma cell-intrinsic trait. Analysis of TCGA patient datasets shows matrisome signature is enriched in lobular carcinomas with overexpression of elastin, collagens, and the collagen modifying enzyme LOXL1. Treatment with the pan LOX inhibitor BAPN and silencing of LOXL1 expression decrease tumor growth, invasion, and metastasis by disrupting ECM structure resulting in decreased ER signaling. We conclude that LOXL1 inhibition is a promising therapeutic strategy for ILC.


Subject(s)
Breast Neoplasms , Carcinoma, Lobular , Amino Acid Oxidoreductases/genetics , Animals , Carcinoma, Lobular/genetics , Extracellular Matrix , Female , Heterografts , Humans , Mice , Receptors, Estrogen
12.
J Cancer Educ ; 25(1): 101-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20082176

ABSTRACT

Breast cancer is a public health issue in numerous countries. Multidisciplinary collaboration is required for patient care, research, and also education of future physicians. This paper uses Kern's framework for curriculum design to demonstrate how a breast diseases module for undergraduate medical students created in 1993 evolved over 15 years. The main outcomes of program refinements were better integrated course content, the development of electronic course documents, and implementation of computer-aided small group learning. A main future challenge is to further develop efficient instructional strategies in line with well-defined learning needs for undergraduate students.


Subject(s)
Breast Neoplasms , Education, Medical, Undergraduate/organization & administration , Interprofessional Relations , Breast Diseases , Curriculum , Female , Humans , Learning , Program Evaluation
13.
Breast Cancer Res ; 9(3): R38, 2007.
Article in English | MEDLINE | ID: mdl-17573968

ABSTRACT

INTRODUCTION: About 70% of breast cancers express oestrogen receptor alpha (ESR1/ERalpha) and are oestrogen-dependent for growth. In contrast with the highly proliferative nature of ERalpha-positive tumour cells, ERalpha-positive cells in normal breast tissue rarely proliferate. Because ERalpha expression is rapidly lost when normal human mammary epithelial cells (HMECs) are grown in vitro, breast cancer models derived from HMECs are ERalpha-negative. Currently only tumour cell lines are available to model ERalpha-positive disease. To create an ERalpha-positive breast cancer model, we have forced normal HMECs derived from reduction mammoplasty tissue to express ERalpha in combination with other relevant breast cancer genes. METHODS: Candidate genes were selected based on breast cancer microarray data and cloned into lentiviral vectors. Primary HMECs prepared from reduction mammoplasty tissue were infected with lentiviral particles. Infected HMECs were characterised by Western blotting, immunofluorescence microscopy, microarray analysis, growth curves, karyotyping and SNP chip analysis. The tumorigenicity of the modified HMECs was tested after orthotopic injection into the inguinal mammary glands of NOD/SCID mice. Cells were marked with a fluorescent protein to allow visualisation in the fat pad. The growth of the graft was analysed by fluorescence microscopy of the mammary glands and pathological analysis of stained tissue sections. Oestrogen dependence of tumour growth was assessed by treatment with the oestrogen antagonist fulvestrant. RESULTS: Microarray analysis of ERalpha-positive tumours reveals that they commonly overexpress the Polycomb-group gene BMI1. Lentiviral transduction with ERalpha, BMI1, TERT and MYC allows primary HMECs to be expanded in vitro in an oestrogen-dependent manner. Orthotopic xenografting of these cells into the mammary glands of NOD/SCID mice results in the formation of ERalpha-positive tumours that metastasise to multiple organs. The cells remain wild type for TP53, diploid and genetically stable. In vivo tumour growth and in vitro proliferation of cells explanted from tumours are dependent on oestrogen. CONCLUSION: We have created a genetically defined model of ERalpha-positive human breast cancer based on normal HMECs that has the potential to model human oestrogen-dependent breast cancer in a mouse and enables the study of mechanisms involved in tumorigenesis and metastasis.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Cell Transformation, Neoplastic , Epithelial Cells/cytology , Estrogens/physiology , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Culture Techniques , Epithelial Cells/pathology , Estrogen Receptor alpha/genetics , Female , Fibroblasts/cytology , Fibroblasts/pathology , Genetic Vectors , Humans , Karyotyping , Lentivirus/genetics , Oligonucleotide Array Sequence Analysis
14.
Rev Med Suisse ; 3(119): 1737-42, 2007 Jul 18.
Article in French | MEDLINE | ID: mdl-17727094

ABSTRACT

Tailoring adjuvant therapy in breast cancer patients relies on prognostic and predictive factors, most of which are currently established by histopathological analysis of tumors. The quality of the assessment of the former (i.e.: tumor size, lymph node status, tumor grade, HER2 status, and lymphovascular invasion) and the latter (estrogen and progesteron receptors expression, HER2 overexpression or amplification) is an essential prerequisite for an optimal therapeutic decision. If the prognostic and predictive values of multigenes signatures are confirmed by on-going clinical studies, this approach could enter the clinical practice in the coming years and result in improved accuracy of adjuvant therapies in breast cancer patients. This approach might especially allow avoiding overtreatment in patients at low risk of recurrence.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Female , Humans , Middle Aged
15.
Oncogene ; 24(29): 4660-71, 2005 Jul 07.
Article in English | MEDLINE | ID: mdl-15897907

ABSTRACT

Previous microarray studies on breast cancer identified multiple tumour classes, of which the most prominent, named luminal and basal, differ in expression of the oestrogen receptor alpha gene (ER). We report here the identification of a group of breast tumours with increased androgen signalling and a 'molecular apocrine' gene expression profile. Tumour samples from 49 patients with large operable or locally advanced breast cancers were tested on Affymetrix U133A gene expression microarrays. Principal components analysis and hierarchical clustering split the tumours into three groups: basal, luminal and a group we call molecular apocrine. All of the molecular apocrine tumours have strong apocrine features on histological examination (P=0.0002). The molecular apocrine group is androgen receptor (AR) positive and contains all of the ER-negative tumours outside the basal group. Kolmogorov-Smirnov testing indicates that oestrogen signalling is most active in the luminal group, and androgen signalling is most active in the molecular apocrine group. ERBB2 amplification is commoner in the molecular apocrine than the other groups. Genes that best split the three groups were identified by Wilcoxon test. Correlation of the average expression profile of these genes in our data with the expression profile of individual tumours in four published breast cancer studies suggest that molecular apocrine tumours represent 8-14% of tumours in these studies. Our data show that it is possible with microarray data to divide mammary tumour cells into three groups based on steroid receptor activity: luminal (ER+ AR+), basal (ER- AR-) and molecular apocrine (ER- AR+).


Subject(s)
Apocrine Glands/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Receptors, Androgen/analysis , Receptors, Androgen/genetics , Receptors, Estrogen/analysis , Receptors, Estrogen/genetics , Biopsy , Breast Neoplasms/classification , Female , Humans , Phenotype , Receptor, ErbB-2/analysis , Receptor, ErbB-2/genetics , Signal Transduction
16.
Sci Rep ; 6: 20277, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26856369

ABSTRACT

Chromogenic immunohistochemistry (IHC) is omnipresent in cancer diagnosis, but has also been criticized for its technical limit in quantifying the level of protein expression on tissue sections, thus potentially masking clinically relevant data. Shifting from qualitative to quantitative, immunofluorescence (IF) has recently gained attention, yet the question of how precisely IF can quantify antigen expression remains unanswered, regarding in particular its technical limitations and applicability to multiple markers. Here we introduce microfluidic precision IF, which accurately quantifies the target expression level in a continuous scale based on microfluidic IF staining of standard tissue sections and low-complexity automated image analysis. We show that the level of HER2 protein expression, as continuously quantified using microfluidic precision IF in 25 breast cancer cases, including several cases with equivocal IHC result, can predict the number of HER2 gene copies as assessed by fluorescence in situ hybridization (FISH). Finally, we demonstrate that the working principle of this technology is not restricted to HER2 but can be extended to other biomarkers. We anticipate that our method has the potential of providing automated, fast and high-quality quantitative in situ biomarker data using low-cost immunofluorescence assays, as increasingly required in the era of individually tailored cancer therapy.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Gene Amplification , Microfluidics/instrumentation , Microfluidics/methods , Receptor, ErbB-2/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Female , Fluorescent Antibody Technique , Humans , In Situ Hybridization, Fluorescence , Pilot Projects , Receptor, ErbB-2/metabolism
17.
Cancer Cell ; 29(3): 407-422, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26947176

ABSTRACT

Seventy-five percent of breast cancers are estrogen receptor α positive (ER⁺). Research on these tumors is hampered by lack of adequate in vivo models; cell line xenografts require non-physiological hormone supplements, and patient-derived xenografts (PDXs) are hard to establish. We show that the traditional grafting of ER⁺ tumor cells into mammary fat pads induces TGFß/SLUG signaling and basal differentiation when they require low SLUG levels to grow in vivo. Grafting into the milk ducts suppresses SLUG; ER⁺ tumor cells develop, like their clinical counterparts, in the presence of physiological hormone levels. Intraductal ER⁺ PDXs are retransplantable, predictive, and appear genomically stable. The model provides opportunities for translational research and the study of physiologically relevant hormone action in breast carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estrogen Receptor alpha/genetics , Mammary Glands, Human/pathology , Tumor Microenvironment/genetics , Animals , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Transforming Growth Factor beta/genetics
18.
J Nucl Med ; 46(3): 395-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15750149

ABSTRACT

UNLABELLED: Previous reports have shown that axillary sentinel lymph node (ASLN) radiodetection allows accurate axillary staging for patients with early breast cancer. Radioguided surgery implies the use of a gamma-probe to count the emitted radioactivity of marked ASLNs. Several gamma-probes are commercially available, each with its own properties. The clinical impact of the type of gamma-probe used for ASLN radiodetection remains to be evaluated. METHODS: Three commercially available gamma-probes were evaluated: a scintillator with a bismuth germanate crystal (probe A), a semiconductor with a cadmium telluride crystal (probe B), and a semiconductor with a cadmium zinc telluride crystal (probe C). Two hundred patients with early breast cancer were prospectively enrolled to undergo ASLN radiodetection and axillary lymphadenectomy. ASLN mapping consisted of injecting (99m)Tc-sulfur-colloid around the tumor. For each patient, sentinel lymph nodes were counted successively with the 3 probes and the sensitivity of each gamma-probe was determined from ASLN residual activity. The results of detection rates and false-negative rates for each probe were compared. RESULTS: Mean residual ASLN activity was 52 kBq (range, 0.07-189 kBq). Sensitivity was compared among the 3 probes and found to be best for probe A. The detection rate of probe A was significantly better than that of probe B (93% vs. 86%, P = 0.05) but not different from that of probe C (93% vs. 90%). No differences in false-negative rates were observed among the 3 probes. CONCLUSION: ASLN detection rate depends on the type of gamma-probe used. Because failure to detect the ASLN leads to complete axillary lymphadenectomy, involving local morbidity and other sequelae, the type of gamma-probe must be considered important for sentinel lymph node radiodetection.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Gamma Cameras , Lymph Nodes/diagnostic imaging , Lymph Nodes/pathology , Sentinel Lymph Node Biopsy/instrumentation , Surgery, Computer-Assisted/instrumentation , Equipment Design , Equipment Failure Analysis , Female , Humans , Lymphatic Metastasis , Middle Aged , Radionuclide Imaging/instrumentation , Radionuclide Imaging/methods , Reproducibility of Results , Sensitivity and Specificity , Sentinel Lymph Node Biopsy/methods , Surgery, Computer-Assisted/methods , Technology Assessment, Biomedical
19.
Ann Pathol ; 23(6): 617-22, 2003 Dec.
Article in French | MEDLINE | ID: mdl-15094603

ABSTRACT

The HER2 proto-oncogene encodes a transmembrane protein, which is considered to function as a growth factor receptor. Overexpression of this protein found by immunohistochemistry in about 20% of infiltrating breast carcinomas, has a predictive value of response to treatment by trastuzumab, an anti-HER2 humanized monoclonal antibody. Search for HER2 gene amplification is necessary to adapt the immunohistochemical technique quality and also in the cases of delicate analysis or weak overexpression. It is usually carried out by Fluorescence In Situ Hybridization (FISH). A more recent hybridization technique, named CISH because of its chromogenic revelation is an alternative method, which gives highly correlated results with FISH. We present details of this technique, which may be more familiar for the pathologists than FISH, because reading analysis is similar to that of immunohistochemical staining.


Subject(s)
Chromogenic Compounds/analysis , Genes, erbB-2 , In Situ Hybridization/methods , Nucleic Acid Amplification Techniques , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/genetics , Chromosomes, Human, Pair 17/genetics , DNA Probes , Digoxigenin/analysis , Female , Humans , In Situ Hybridization, Fluorescence , Proto-Oncogene Mas , Specimen Handling
20.
Crit Rev Oncol Hematol ; 92(3): 235-57, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25129506

ABSTRACT

The WHO classification of breast tumors distinguishes, besides invasive breast cancer 'of no special type' (former invasive ductal carcinoma, representing 60-70% of all breast cancers), 30 special types, of which invasive lobular carcinoma (ILC) is the most common (5-15%). We review the literature on (i) the specificity and heterogeneity of ILC biology as documented by various analytical techniques, including the results of molecular testing for risk of recurrence; (ii) the impact of lobular histology on prediction of prognosis and effect of systemic therapies in patients. Though it is generally admitted that ILC has a better prognosis than IDC, is endocrine responsive, and responds poorly to chemotherapy, currently available data do not unanimously support these assumptions. This review demonstrates some lack of specific data and a need for improving clinical research design to allow oncologists to make informed systemic therapy decisions in patients with ILC. Importantly, future studies should compare various endpoints in ILC breast cancer patients among the group of hormonosensitive breast cancer.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Carcinoma, Lobular/diagnosis , Carcinoma, Lobular/pathology , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Carcinoma, Lobular/mortality , Carcinoma, Lobular/therapy , Female , Genetic Testing , Genomics , Humans , Neoplasm Grading , Neoplasm Invasiveness , Prognosis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL