Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Andrology ; 7(4): 463-468, 2019 07.
Article in English | MEDLINE | ID: mdl-30786164

ABSTRACT

BACKGROUND: The microRNA-371a-3p (miR-371a-3p) has been reported to be an informative liquid biopsy (serum and plasma) molecular biomarker for both diagnosis and follow-up of patients with a malignant (testicular) germ cell tumor ((T)GCT). It is expressed in all histological cancer elements, with the exception of mature teratoma. However, normal testis, semen, and serum of males with a disrupted testicular integrity without a TGCT may contain miR-371a-3p levels above threshold, of which the cellular origin is unknown. OBJECTIVES: Therefore, a series of relevant tissues (frozen and formalin-fixed paraffin-embedded (FFPE), when available) from the complete male urogenital tract (i.e., kidney to urethra and testis to urethra) and semen was investigated for miR-371a-3p levels using targeted quantitative RT-PCR (qRT-PCR). MATERIALS AND METHODS: In total, semen of males with normospermia (n = 11) and oligospermia (n = 3) was investigated, as well as 88 samples derived from 32 different patients. The samples represented one set of tissues related to the entire male urogenital tract (11 anatomical locations), three sets for 10 locations, and four sets for six locations. RESULTS: All testis parenchyma (n = 17) cases showed low miR-371a-3p levels. Eight out of 14 (57%) semen samples showed detectable miR-371a-3p levels, irrespective of the amount of motile spermatozoa, but related to sperm concentration and matched Johnsen score (Spearman's rho correlation coefficient 0.849 and 0.871, p = 0.000, respectively). In all other tissues investigated, miR-371a-3p could not be detected. DISCUSSION: This study demonstrates that the miR-371a-3p in healthy adult males is solely derived from the germ cell compartment. CONCLUSIONS: The observation is important in the context of applying miR-371a-3p as molecular liquid biopsy biomarker for diagnosis and follow-up of patients with malignant (T)GCT. Moreover, miR-371a-3p might be an informative seminal biomarker for testicular germ cell composition.


Subject(s)
Genitalia, Male/metabolism , MicroRNAs/metabolism , Semen/metabolism , Urinary Tract/metabolism , Humans , Male , Oligospermia/metabolism , Reference Values
2.
Andrology ; 3(1): 85-91, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25382245

ABSTRACT

microRNAs (miRs) are short non-coding RNA molecules (≈21 nucleotides) involved in regulation of translation. As such they are crucial for normal cell development and differentiation as well as cellular maintenance. Dysregulation of miRs has been reported in various diseases, including cancer. Interestingly, miRs can be informative as tumor classifiers and disease biomarkers. Recent studies demonstrated the presence of miRs in body fluids like serum, thus providing a putative non-invasive tool to study and monitor disease state. Earlier targeted studies by several independent groups identified specific embryonic miRs as characteristic for germ cell tumors (GCT) (miR-371-2-3 & miR-302/367 clusters). This study reports a high-throughput miR profiling (≈750 miRs) approach on serum from testicular germ cell tumor patients (14 seminoma and 10 non-seminoma) and controls (n = 11), aiming at independent identification of miRs as candidate biomarkers for testicular GCT. A magnetic bead capture system was used to isolate miRs from serum. Subsequently, the TaqMan Array Card 3.0 platform was used for profiling. The previously identified miRs 371 and 372 were confirmed to be specifically elevated in serum from germ cell tumor patients. In addition, several novels miRs were identified that were discriminative between germ cell cancer and controls: miR-511, -26b, -769, -23a, -106b, -365, -598, -340, and let-7a. In conclusion, this study validates the power of the embryonic miRs 371 and 372 in detecting malignant GCT (SE and NS) based on serum miR levels and identifies several potential novel miR targets.


Subject(s)
Biomarkers, Tumor/blood , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , MicroRNAs/blood , Neoplasms, Germ Cell and Embryonal/blood , Seminoma/blood , Testicular Neoplasms/blood , Base Sequence , Biomarkers, Tumor/genetics , Case-Control Studies , Discriminant Analysis , Humans , Male , MicroRNAs/genetics , Molecular Sequence Data , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Predictive Value of Tests , Seminoma/genetics , Seminoma/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/pathology
3.
Br J Cancer ; 107(6): 947-55, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22892392

ABSTRACT

BACKGROUND: High BCAR4 and ERBB2 mRNA levels in primary breast cancer associate with tamoxifen resistance and poor patient outcome. We determined whether BCAR4 expression sensitises breast cancer cells to lapatinib, and identifies a subgroup of patients who possibly may benefit from ERBB2-targeted therapies despite having tumours with low ERBB2 expression. METHODS: Proliferation assays were applied to determine the effect of BCAR4 expression on lapatinib treatment. Changes in cell signalling were quantified with reverse-phase protein microarrays. Quantitative reverse-transcriptase polymerase chain reaction (RT-PCR) of ERBB2 and BCAR4 was performed in 1418 primary breast cancers. Combined BCAR4 and ERBB2 mRNA levels were evaluated for association with progression-free survival (PFS) in 293 oestrogen receptor-α (ER)-positive patients receiving tamoxifen as first-line monotherapy for recurrent disease. RESULTS: BCAR4 expression strongly sensitised ZR-75-1 and MCF7 breast cancer cells to the combination of lapatinib and antioestrogens. Lapatinib interfered with phosphorylation of ERBB2 and its downstream mediators AKT, FAK, SHC, STAT5, and STAT6. Reverse transcriptase-PCR analysis showed that 27.6% of the breast cancers were positive for BCAR4 and 22% expressed also low levels of ERBB2. The clinical significance of combining BCAR4 and ERBB2 mRNA status was underscored by the finding that the group of patients having BCAR4-positive/ERBB2-low-expressing cancers had a shorter PFS on tamoxifen treatment than the BCAR4-negative group. CONCLUSION: This study shows that BCAR4 expression identifies a subgroup of ER-positive breast cancer patients without overexpression of ERBB2 who have a poor outcome and might benefit from combined ERBB2-targeted and antioestrogen therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estrogen Receptor Modulators/therapeutic use , Molecular Targeted Therapy/methods , Quinazolines/therapeutic use , RNA, Untranslated/metabolism , Receptor, ErbB-2/metabolism , Tamoxifen/therapeutic use , Adult , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Tumor/analysis , Breast Neoplasms/mortality , Cell Proliferation , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lapatinib , Middle Aged , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Quinazolines/pharmacology , RNA, Long Noncoding , RNA, Messenger/metabolism , RNA, Untranslated/drug effects , RNA, Untranslated/genetics , Receptor, ErbB-2/drug effects , Receptor, ErbB-2/genetics , Receptors, Estrogen/analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
4.
Br J Cancer ; 103(8): 1284-91, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20859285

ABSTRACT

BACKGROUND: Breast cancer anti-oestrogen resistance 4 (BCAR4) was identified in a search for genes involved in anti-oestrogen resistance in breast cancer. We explored whether BCAR4 is predictive for tamoxifen resistance and prognostic for tumour aggressiveness, and studied its function. METHODS: BCAR4 mRNA levels were measured in primary breast tumours, and evaluated for association with progression-free survival (PFS) and clinical benefit in patients with oestrogen receptor (ERα)-positive tumours receiving tamoxifen as first-line monotherapy for advanced disease. In a separate cohort of patients with lymph node-negative, ERα-positive cancer, and not receiving systemic adjuvant therapy, BCAR4 levels were evaluated for association with distant metastasis-free survival (MFS). The function of BCAR4 was studied with immunoblotting and RNA interference in a cell model. RESULTS: Multivariate analyses established high BCAR4 mRNA levels as an independent predictive factor for poor PFS after start of tamoxifen therapy for recurrent disease. High BCAR4 mRNA levels were associated with poor MFS and overall survival, reflecting tumour aggressiveness. In BCAR4-expressing cells, phosphorylation of v-erb-b2 erythroblastic leukaemia viral oncogene homolog (ERBB)2, ERBB3, and their downstream mediators extracellular signal-regulated kinase 1/2 and v-akt murine thymoma viral oncogene homolog (AKT) 1/2, was increased. Selective knockdown of ERBB2 or ERBB3 inhibited proliferation, confirming their role in BCAR4-induced tamoxifen resistance. CONCLUSION: BCAR4 may have clinical relevance for tumour aggressiveness and tamoxifen resistance. Our cell model suggests that BCAR4-positive breast tumours are driven by ERBB2/ERBB3 signalling. Patients with such tumours may benefit from ERBB-targeted therapy.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma/drug therapy , Carcinoma/pathology , Crk-Associated Substrate Protein/physiology , Drug Resistance, Neoplasm/genetics , Tamoxifen/therapeutic use , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , Adult , Aged , Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Pharmacological/analysis , Biomarkers, Pharmacological/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Carcinoma/genetics , Carcinoma/mortality , Cell Line, Tumor , Crk-Associated Substrate Protein/genetics , Crk-Associated Substrate Protein/metabolism , Female , Humans , Middle Aged , Neoplasm Invasiveness , RNA, Long Noncoding , RNA, Untranslated , Retrospective Studies , Survival Analysis
5.
Br J Cancer ; 101(11): 1824-32, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19904269

ABSTRACT

BACKGROUND: Endocrine therapies of breast cancer are effective but ultimately fail because of the development of treatment resistance. We have previously revealed several genes leading to tamoxifen resistance in vitro by retroviral insertion mutagenesis. To understand the manner in which these genes yield tamoxifen resistance, their effects on global gene expression were studied and those genes resulting in a distinct gene expression profile were further investigated for their clinical relevance. METHODS: Gene expression profiles of 69 human breast cancer cell lines that were made tamoxifen resistant through retroviral insertion mutagenesis were obtained using oligonucleotide arrays and analysed with bioinformatic tools. mRNA levels of NCOR2 and CITED2 in oestrogen receptor-positive breast tumours were determined by quantitative RT-PCR. mRNA levels were evaluated for association with metastasis-free survival (MFS) in 620 patients with lymph node-negative primary breast cancer who did not receive systemic adjuvant therapy, and with clinical benefit in 296 patients receiving tamoxifen therapy for recurrent breast cancer. RESULTS: mRNA expression profiles of most tamoxifen-resistant cell lines were strikingly similar, except for the subgroups of cell lines in which NCOR2 or CITED2 were targeted by the retrovirus. Both NCOR2 and CITED2 mRNA levels were associated with MFS, that is, tumour aggressiveness, independently of traditional prognostic factors. In addition, high CITED2 mRNA levels were predictive for a clinical benefit from first-line tamoxifen treatment in patients with advanced disease. CONCLUSIONS: Most retrovirally targeted genes yielding tamoxifen resistance in our cell lines do not impose a distinctive expression profile, suggesting that their causative role in cell growth may be accomplished by post-transcriptional processes. The associations of NCOR2 and CITED2 with outcome in oestrogen receptor-positive breast cancer patients underscore the clinical relevance of functional genetic screens to better understand disease progression, which may ultimately lead to the development of improved treatment options.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Antagonists/pharmacology , Nuclear Receptor Co-Repressor 2/metabolism , Repressor Proteins/metabolism , Tamoxifen/pharmacology , Trans-Activators/metabolism , Adult , Aged , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Humans , Lymphatic Metastasis , Middle Aged , Nuclear Receptor Co-Repressor 2/biosynthesis , Nuclear Receptor Co-Repressor 2/genetics , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Trans-Activators/biosynthesis , Trans-Activators/genetics
6.
Drugs ; 61(12): 1721-33, 2001.
Article in English | MEDLINE | ID: mdl-11693462

ABSTRACT

Tamoxifen has been used for the systemic treatment of patients with breast cancer for nearly three decades. Treatment success is primarily dependent on the presence of the estrogen receptor (ER) in the breast carcinoma. While about half of patients with advanced ER-positive disease immediately fail to respond to tamoxifen, in the responding patients the disease ultimately progresses to a resistant phenotype. The possible causes for intrinsic and acquired resistance have been attributed to the pharmacology of tamoxifen, alterations in the structure and function of the ER, the interactions with the tumour environment and genetic alterations in the tumour cells. So far no prominent mechanism leading to resistance has been identified. The recent results of a functional screen for breast cancer antiestrogen resis- tance (BCAR) genes responsible for development of tamoxifen resistance in human breast cancer cells are reviewed. Individual BCAR genes can transform estrogen-dependent breast cancer cells into estrogen-independent and tamoxifen-resistant cells in vitro. Furthermore, high levels of BCAR1/pl30Cas protein in ER-positive primary breast tumours are associated with intrinsic resistance to tamoxifen treatment. These results indicate a prominent role for alternative growth control pathways independent of ER signalling in intrinsic tamoxifen resistance of ER-positive breast carcinomas. Deciphering the differentiation characteristics of normal and malignant breast epithelial cells with respect to proliferation control and regulation of cell death (apoptosis) is essential for understanding therapy response and development of resistance of breast carcinoma.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Tamoxifen/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estrogen Antagonists/pharmacology , Female , Humans , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Receptors, Estrogen/physiology
7.
EMBO J ; 17(10): 2799-808, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9582273

ABSTRACT

The antiestrogen tamoxifen is important in the treatment of hormone-dependent breast cancer, although development of resistance is inevitable. To unravel the molecular mechanisms of antiestrogen resistance, a search for involved genes was initiated. Retrovirus-mediated insertional mutagenesis was applied to human ZR-75-1 breast cancer cells. Infected cells were subjected to tamoxifen selection and a panel of resistant cell clones was established. Screening for a common integration site resulted in the identification of a novel gene designated BCAR3. Transfer of this locus by cell fusion or transfection of the BCAR3 cDNA to ZR75-1 and MCF-7 cells induces antiestrogen resistance. BCAR3 represents a putative SH2 domain-containing protein and is partly homologous to the cell division cycle protein CDC48.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/genetics , Estrogen Antagonists/pharmacology , Receptors, Cyclic AMP/genetics , Tamoxifen/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Breast Neoplasms/drug therapy , Cell Fusion , Cloning, Molecular , DNA, Complementary , DNA, Neoplasm , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Humans , Molecular Sequence Data , Protein Biosynthesis , RNA, Messenger , Sequence Homology, Amino Acid , Transcription, Genetic , Tumor Cells, Cultured
8.
Pathol Res Pract ; 192(7): 743-51, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8880875

ABSTRACT

Endocrine therapy is effective in the treatment of breast cancer. Adjuvant treatment with tamoxifen reduces tumor recurrence and achieves increased survival. In metastatic disease, tamoxifen treatment accomplishes objective responses in +/- 50% of the patients with estrogen receptor-positive primary tumors. However, the response duration is limited due to the inevitable development of metastases resistant to tamoxifen. The mechanisms leading to tamoxifen resistance are largely unknown. We have set out to identify genetic pathways in the tumor cells causing failure of tamoxifen therapy. We selected an estrogen-dependent human breast cancer cell line (ZR-75-1) and demonstrated that genetic and epigenetic alterations can change the hormone-response phenotype of these cells. Subsequently, we applied insertional mutagenesis with defective retroviruses to these ZR-75-1 breast cancer cells. Integration of a retrovirus in the cellular DNA alters the genome structure and may modify the expression of genes in its surroundings. As a result of the altered gene expression, the biological phenotype of the infected cell may be changed. The infected ZR-75-1 cells were subjected to tamoxifen selection and a panel of tamoxifen-resistant cell lines has been established. Screening for a common integration site for the retrovirus has provided, so far, compelling evidence for the involvement of at least one genetic locus (BCAR 1) in breast cancer antiestrogen resistance in vitro.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Estrogens/physiology , Female , Humans
9.
Int J Cancer ; 63(6): 790-3, 1995 Dec 11.
Article in English | MEDLINE | ID: mdl-8847135

ABSTRACT

The prognostic value of epidermal growth factor receptor (EGFR) expression and its biological role in estrogen receptor-positive (ER+) and ER-negative (ER-) primary breast cancer is controversial. In this study, distributions of ER, progesterone receptor and EGFR have been established using immunohistochemistry in both primary breast tumors and their matched axillary lymph node metastases of 26 patients or their matched distant metastases of 2 patients. In addition, 5 patients with bilateral breast cancer were studied. ER+ tumor cells were detected in 22 (69%) and EGFR+ tumor cells were detected in 11 (34%) primary breast carcinomas. Expression of ER and EGFR was inverse regarding the individual tumor cells in both primary tumors and metastases. Relationship of EGFR expression with poorly differentiated and large breast tumors was observed. Furthermore, primary tumors with a predominant lobular component were ER+ and, with one exception, EGFR-. Invasive ductal carcinomas were more frequently EGFR+. No apparent differences in receptor expression were observed between primary tumors and lymph node metastases or chronously or metachronously occurring bilateral breast cancers. Only one ER+ primary tumor showed a switch to EGFR expression in the involved lymph node. Our study shows that a shift in receptor phenotype between primary tumors and lymph node metastases is a rare event and, thus, additional analyses of involved lymph nodes will not likely serve as a better predictor for response to anti-estrogen therapy. We conclude that expression of EGFR is not a prerequisite for development of metastases.


Subject(s)
Breast Neoplasms/metabolism , ErbB Receptors/biosynthesis , Estrogens/biosynthesis , Lymph Nodes/metabolism , Progesterone/biosynthesis , Adult , Aged , Breast Neoplasms/pathology , Female , Humans , Lymphatic Metastasis , Middle Aged
10.
Mol Endocrinol ; 8(11): 1474-83, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7533260

ABSTRACT

Antagonists of steroid hormones are clinically important in the management of breast cancer. However, the duration of response is limited due to the development of hormone-independent tumors in virtually all cases. In an attempt to obtain insight into the mechanisms underlying antiestrogen resistance, the consequences of epigenetic changes in gene expression were studied in vitro. Estrogen-dependent ZR-75-1 human breast cancer cells were treated with 5-azacytidine, an inhibitor of DNA methylation, and cultured in the absence of estradiol or in the presence of antiestrogens. Estrogen-independent cell colonies developed within 3 weeks at high frequency in 5-azacytidine-treated cultures (0.7 x 10(-3), in contrast to control cultures (< or = 10(-8). The derived cells (ZR/AZA) were resistant to 4-hydroxytamoxifen and ICI 164,384, independent of the selection protocol, but had lost the ability to grow anchorage-independent. Whereas expression of estrogen receptor, progesterone receptor, and pS2 were down-regulated, expression of epidermal growth factor (EGF) receptor and HER2/neu were increased in ZR/AZA cells. In contrast to the stable altered expression patterns of estrogen receptor and EGF receptor, transient keratin 7 expression was observed. Transforming growth factor-alpha mRNA was identified in ZR-75-1 cells and ZR/AZA cells and EGF-like peptides were secreted in the culture medium. Proliferation of ZR/AZA cells could be partially inhibited with an EGF receptor-blocking antibody. Presence of both growth factor receptors and possible ligands suggests the development of an autocrine growth mechanism. Our data show that epigenetic alterations of gene expression result in rapid progression of breast cancer cells to hormone independence.


Subject(s)
Breast Neoplasms/secondary , Estrogens/physiology , Neoplasms, Hormone-Dependent/genetics , Proteins , Receptors, Estradiol/metabolism , Azacitidine/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Down-Regulation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Keratins/biosynthesis , Neoplasm Proteins/biosynthesis , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Polyunsaturated Alkamides , Receptor, ErbB-2/metabolism , Receptors, Progesterone/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Trefoil Factor-1 , Tumor Cells, Cultured , Tumor Suppressor Proteins
11.
Am J Pathol ; 144(6): 1238-46, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7515559

ABSTRACT

Distribution of estrogen (ER), progesterone (PR) receptors, and epidermal growth factor (EGF) receptors was assayed by dual staining immunohistochemistry on 28 selected cytosolic ER-positive breast carcinomas and 22 nonmalignant breast tissues. ER-positive tumor cells were detected in 26 (93%) and EGF receptor positive tumor cells were detected in 7 (25%) carcinomas. In five tumors both ER and EGF receptors were detected but localized in distinct tumor cells. Only in one case of ductal carcinoma in situ co-expression was observed in a subset of tumor cells. In contrast, simultaneous expression of ER/PR and EGF receptors was observed in non-neoplastic ductal remnants in the majority of the carcinomas and the fibroadenomas. In addition, double-positive cells were occasionally detected in luminal epithelial cells of normal breast tissue and mastopathies. This study shows that ER/PR and EGF receptors in breast tumor cells are inversely related at the single cell level. However, demonstration of ER/PR and EGF receptors in individual normal luminal cells shows that expression is not mutually exclusive.


Subject(s)
Breast Neoplasms/chemistry , Breast/chemistry , Carcinoma, Ductal, Breast/chemistry , ErbB Receptors/analysis , Fibroadenoma/chemistry , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Breast/pathology , Breast/ultrastructure , Breast Neoplasms/pathology , Breast Neoplasms/ultrastructure , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/ultrastructure , Cell Line , Fibroadenoma/pathology , Fibroadenoma/ultrastructure , Humans , Immunohistochemistry/methods , Staining and Labeling , Tumor Cells, Cultured
12.
Histochem J ; 26(4): 306-10, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7518813

ABSTRACT

A new dual immunocytochemical assay for oestrogen receptor (ER) and epidermal growth factor receptor (EGFR) has been developed. It has been tested in a variety of conditions using cell culture lines and the results correlate well with those obtained from single immunocytochemical assays. MCF-7 and A431 cells and a mixture of the two types of cell were assessed immunocytochemically for ER and EGFR. ER showed immunopositivity of 30% in MCF-7 cells, 10% in the mixture and 0% in A431 cells. EGFR demonstrated immunopositivity of 0% in MCF-7 cells, 70% in the mixture and 100% in A431 cells. Dual immunocytochemical assays using anti-ER followed by anti-EGFR monoclonal antibodies on single histological sections showed similar reactivity to the single assays. Three staining patterns were seen in the mixture: ER+/EGFR- (MCF-7 cells), ER-/EGFR- (MCF-7 cells) and ER-/EGFR+ (A431 cells). ZR-75-1 and MDA-MB-231 cells and their retrovirally transfected counterparts ZR/HERc and MDA/HEGO cells were then analysed. The dual assay revealed the fourth phenotype (ER+/EGFR+) in 40% of ZR/HERc cells and in 10% of MDA/HEGO cells. This is the first description of a dual immunocytochemical assay system for ER and EGFR on single 5 microns frozen section samples. Studies are now underway assessing breast carcinoma sections which may allow investigation of the clonality of human breast cancer.


Subject(s)
Breast Neoplasms/chemistry , Carcinoma, Squamous Cell/chemistry , ErbB Receptors/analysis , Receptors, Estrogen/analysis , Frozen Sections , Humans , Immunohistochemistry , Staining and Labeling , Tumor Cells, Cultured
13.
Mol Endocrinol ; 7(7): 870-8, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8413311

ABSTRACT

Duration of response to antiestrogen therapy in metastatic breast cancer is limited due to the development of antiestrogen-resistant tumors. The mechanisms involved are not understood but could originate from (epi)genetic alterations within the tumor cells. We have applied in vitro random insertional mutagenesis with replication defective retroviruses to identify those genes playing a key role in development of antiestrogen resistance in human breast cancer cells. Eighty antiestrogen-resistant cell clones were isolated from 7 x 10(8) estrogen-dependent ZR-75-1 cells, mass-infected with defective retroviruses and subjected to 4-OH-tamoxifen selection. Integration site-specific DNA probes were made by inverse polymerase chain reaction techniques and used to search for common integration sites. Six cell clones were identified with retroviral genome integrations in the same orientation in a single locus, designated breast cancer antiestrogen resistance locus-1 (bcar-1). These bcar-1 cell clones had lost estrogen receptor expression and had become estrogen independent. Our results strongly suggest that alteration of the bcar-1 locus is responsible for development of antiestrogen resistance in human breast cancer cells in vitro. In addition, we have shown that in vitro insertional mutagenesis using defective retroviruses can be applied for gene tagging in human cells.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA Transposable Elements/genetics , DNA, Neoplasm/genetics , DNA, Viral/genetics , Estrogen Antagonists/pharmacology , Retroviridae/genetics , Virus Integration , Autoradiography , Base Sequence , Blotting, Southern , Chromosome Mapping , DNA Probes , Drug Resistance , Humans , Molecular Sequence Data , Mutagenesis, Insertional , Polymerase Chain Reaction , Tamoxifen/pharmacology , Tumor Cells, Cultured , Virus Integration/genetics
15.
Cancer Res ; 52(18): 5082-8, 1992 Sep 15.
Article in English | MEDLINE | ID: mdl-1516065

ABSTRACT

Epidermal growth factor (EGF) receptor is inversely related to expression of estrogen receptor (ER) and progesterone receptor in primary breast tumors and is a negative predictor for response to endocrine therapy. To investigate a possible causal role of EGF receptor expression in breast cancer progression to hormone independence, we have created an experimental cell system. Epidermal growth factor receptor complementary DNA was introduced in estrogen-dependent ZR-75-1 breast cancer cells, and the resulting ZR/HERc cells exhibited a mitogenic response to epidermal growth factor, thus bypassing estrogen dependence. This EGF-induced proliferation could not be inhibited by antiestrogens. In addition, we noted changes in cell morphology and keratin expression of EGF-stimulated ZR/HERc cells, suggestive of an altered differentiation state. Furthermore, intolerance of functional ER and EGF receptor signal transduction pathways in ZR/HERc cells was observed during simultaneous activation, which possibly explains the inverse relationship of ER and EGF receptor expression in primary tumors. In contrast to the parental cells, ZR/HERc cells rapidly progressed to a stable ER-negative phenotype when cultured in the presence of the antiestrogen hydroxy-tamoxifen. These results suggest a possible role for EGF receptor in progression of breast cancer to hormone independence.


Subject(s)
Breast Neoplasms/pathology , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Estrogens/physiology , Cell Differentiation/drug effects , Cell Division/drug effects , Drug Resistance , ErbB Receptors/genetics , Gene Expression , In Vitro Techniques , Receptors, Estradiol/physiology , Signal Transduction , Tamoxifen/pharmacology , Transfection , Tumor Cells, Cultured
16.
Blood ; 79(11): 2867-72, 1992 Jun 01.
Article in English | MEDLINE | ID: mdl-1586734

ABSTRACT

To create experimental systems that facilitate studies aimed at the responsiveness of hematopoietic progenitors to interleukin (IL)-6 in combination with IL-3, we introduced the human IL-6 receptor (hIL-6R) into the IL-3-dependent cell line 32D. For this purpose, a retroviral vector containing the hIL-6R cDNA was constructed. 32D parental cells did not respond to IL-6, neither alone nor in combination with increasing concentrations of IL-3, and did not express detectable numbers of IL-6R as determined by 125I-IL-6 binding. 32D/hIL-6R cells expressed high-affinity IL-6 binding and responded synergistically to IL-6 in combination with suboptimal amounts of IL-3 in DNA synthesis assays. In addition, IL-6 promoted the short-term survival of IL-3-responsive clonogenic 32D/hIL-6R cells. On the other hand, although introduction of hIL-6R resulted in the formation of high-affinity IL-6 receptor structures in the IL-2-dependent thymocyte cell line CTLL, CTLL/hIL-6R cells did not respond to IL-6 in synergy with IL-2. We conclude that 32D cells possess the intracellular machinery permissive for IL-6 signal transduction. Murine IL-3-dependent cell lines with ectopic IL-6 receptors can serve as models for dissecting the molecular basis of IL-6 responses in primitive hematopoietic cells.


Subject(s)
Hematopoietic Stem Cells/metabolism , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Receptors, Immunologic/physiology , Animals , Cell Division , Cell Line , Cell Survival , DNA/biosynthesis , DNA/genetics , Drug Synergism , Gene Expression , Hematopoietic Stem Cells/cytology , Humans , Mice , Receptors, Immunologic/genetics , Receptors, Interleukin-6 , Transfection
18.
Blood ; 75(11): 2097-101, 1990 Jun 01.
Article in English | MEDLINE | ID: mdl-2189505

ABSTRACT

We investigated the proliferation-inducing effects of human recombinant interleukin-7 (IL-7) on acute lymphoblastic leukemia (ALL) cells. It is shown that IL-7 stimulates DNA synthesis in ALL cells of B-cell precursor (n = 5) as well as immature T-cell origin (n = 2). Cytogenetic analysis of the cells of four patients proliferating in IL7-supplemented cultures established the leukemic descendence of the IL-7-responsive cells. 125I-IL-7 binding experiments with the cells of one patient and with two ALL cell lines showed the presence of two types of IL-7 receptors: one with a high affinity (kd 29 to 51 pmol/L) and one with a low affinity (kd 2.3 to 76 nmol/L) for the ligand. We conclude that IL-7 is one of the cytokines involved in the complex regulation of ALL cell proliferation.


Subject(s)
Burkitt Lymphoma/pathology , Growth Substances/pharmacology , Interleukin-7/pharmacology , Leukemia, Lymphoid/pathology , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , B-Lymphocytes/ultrastructure , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Female , Fluorescent Antibody Technique , Humans , Interleukin-7/metabolism , Male , Receptors, Immunologic/metabolism , Receptors, Interleukin-7 , Stem Cells/drug effects , Stem Cells/pathology , Stem Cells/ultrastructure , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , T-Lymphocytes/ultrastructure , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
19.
Leukemia ; 3(5): 356-62, 1989 May.
Article in English | MEDLINE | ID: mdl-2497281

ABSTRACT

We have tested a panel of recombinant hematopoietic growth factors (HGF) including the interleukins (IL) 1, 2, 3, 4, and 6 and the colony stimulating factors GM-CSF, G-CSF, M-CSF for their ability to induce proliferation of precursor B acute lymphoblastic leukemia cells (ALL) from 19 patients. In Ficoll-Isopaque isolated and T cell-depleted ALL bone marrow samples, IL2 (two cases), IL3 (four cases), and GM-CSF (one case) infrequently stimulated DNA synthesis measured by 3H-thymidine (TdR) uptake, and the other recombinant growth factors completely failed to do so. In repeat experiments with ALL blasts purified by fluorescence activated cell sorting (FACS), IL2, IL3, and GM-CSF responses could not be reproduced, suggesting that nonleukemic contaminant cells, and not the ALL blasts, had been stimulated by these factors. Cocktails containing combinations of IL1-IL4 and IL6 also lacked proliferation inducing potency. Depending on the purity of the incubated ALL cell samples, an impure preparation of B cell growth factors that has been reported to contain a highly effective stimulatory activity for precursor B ALL cells induced proliferation of residual normal cells as well as the ALL cells, as was evident from combined analysis of DNA synthesis and karyotyping. Exposure of the ALL blasts to artificial activators of protein kinase C and Ca2+ mobilization resulted in significant rises in 3H-TdR uptake, suggesting that these intracellular compounds are involved in transducing signals that upregulate proliferation. Although it remains possible that some of the human recombinant growth factors promote the growth of precursor B ALL cells in combination with other stimuli, a dominant role in the regulation of proliferation of these cells cannot be attributed to any of these cytokines at the present time.


Subject(s)
Colony-Stimulating Factors/pharmacology , Interleukins/pharmacology , Leukemia, Lymphoid/pathology , Neoplastic Stem Cells/pathology , B-Lymphocytes , Calcimycin/pharmacology , Cell Division/drug effects , Humans , Interleukin-4 , Recombinant Proteins/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
20.
Leuk Res ; 13(11): 973-80, 1989.
Article in English | MEDLINE | ID: mdl-2481793

ABSTRACT

Recent studies have suggested that interleukin-2 (IL-2), tumor necrosis factor (TNF) and a crude preparation of B-cell growth factors (BCGF) have a regulatory role in the proliferation of B-CLL cells. However, interpretation of the experimental data has been complicated by potential methodological pitfalls. To establish the role of these factors in B-CLL growth, we investigated their stimulatory effects under accurately defined in vitro conditions. Purified B-CLL cells were obtained by fluorescence activated cell sorting on the basis of coexpression of CD5 and CD19/CD20/CD24 surface antigens in order to avoid interference of normal cell contamination. Subsequently, the cells were cultured free of serum. TNFs alpha and beta and BCGF induced DNA synthesis in the purified B-CLL cells in five out of six cases, as assessed by 3H-thymidine (TdR) uptake on days 3 and 6 of culture. The stimulating activity of BCGF could be suppressed by the addition of specific anti-TNF monoclonal antibodies, indicating that the BCGF activity had been, to a major extent, due to the presence of TNFs in this impure preparation. IL-2, when added as a single stimulus, induced DNA synthesis in the B-CLL cells in three out of six cases on day 3, but not on day 6 of culture. In some patients, cooperative effects of IL-2 and TNF were observed. Cytogenetic analysis was applied to confirm the CLL origin of the proliferating cells. The phorbol ester TPA or anti-IgM were not required for induction of DNA synthesis in the CLL cells, although TPA potentiated 3H-TdR uptake in three out of six cases, and anti-IgM in one case. Our data demonstrate that IL-2 and TNFs alpha and beta can act as growth factors for B-CLL cells under fully defined in vitro conditions, essentially without the need of TPA or anti-IgM as primary activation signals.


Subject(s)
B-Lymphocytes/pathology , Interleukin-2/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphotoxin-alpha/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Antigens, CD/analysis , Antigens, CD19 , Antigens, Differentiation/analysis , Antigens, Differentiation, B-Lymphocyte/analysis , CD5 Antigens , Cell Division/drug effects , Humans , Immunologic Techniques , Interleukin-4/pharmacology , Karyotyping , Receptors, Antigen, B-Cell/physiology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...