Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Cell Mol Med ; 23(2): 1128-1136, 2019 02.
Article in English | MEDLINE | ID: mdl-30536846

ABSTRACT

Germ cell tumours predominantly of the testis ((T)GCTs) are remarkably chemotherapy sensitive. However, a small proportion of patients fail to be cured with cisplatin-based combination chemotherapy. miR-371a-3p is a new liquid biopsy biomarker for (T)GCTs. The aim of this study was to evaluate clinical utility of plasma miR-371a-3p level in patients starting systemic chemotherapy. Patients were included before the first cycle (N = 180) and second cycle (N = 101) of systemic first line chemotherapy, treated between July 2010 and May 2017. Plasma miR-371a-3p levels were measured with the ampTSmiR test and compared to disease characteristics and outcome. Pretreatment plasma miR-371a-3p levels were increased in 51.7% of cases and associated with number of metastatic sites, presence of lung, retroperitoneal, and mediastinal lymph node metastases, S - stage, IGCCCG risk group, and response to therapy. Patients with a negative pretreatment plasma level had better progression-free survival (PFS) and overall survival (OS) compared to patients being positive for miR-371a-3p (hazard ratio [HR] = 0.26, 95% confidence interval [CI] 0.09-0.71, P = 0.02 for PFS and HR = 0.21, 95% CI 0.07-0.67, P = 0.03 for OS, respectively). Patients negative for miR-371a-3p in both samples had a superior PFS (HR = 0.10, 95% CI 0.01-21.49, P = 0.02) and OS (HR = 0.08, 95% CI 0.01-27.81, P = 0.008) compared to patients with miR-371a-3p positive in both samples (multivariate analyses were non-significant). In total 68% of the patients were S0. This study demonstrates clinical value of plasma miR-371a-3p level in chemotherapy naïve (T)GCT patients starting first line of chemotherapy to predict prognosis.


Subject(s)
MicroRNAs/blood , Neoplasms, Germ Cell and Embryonal/blood , Neoplasms, Germ Cell and Embryonal/genetics , Testicular Neoplasms/blood , Testicular Neoplasms/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Cisplatin/therapeutic use , Female , Humans , Lymphatic Metastasis/drug therapy , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Middle Aged , Neoplasms, Germ Cell and Embryonal/drug therapy , Neoplasms, Germ Cell and Embryonal/pathology , Prognosis , Progression-Free Survival , Retrospective Studies , Testicular Neoplasms/drug therapy , Testicular Neoplasms/pathology , Testis/drug effects , Testis/pathology , Young Adult
2.
J Urol ; 200(1): 126-135, 2018 07.
Article in English | MEDLINE | ID: mdl-29474847

ABSTRACT

PURPOSE: Retroperitoneal lymph node dissection is recommended for residual masses greater than 1 cm after chemotherapy of nonseminomatous germ cell tumors. Currently there is no reliable predictor of post-chemotherapy retroperitoneal lymph node dissection histology. Up to 50% of patients harbor necrosis/fibrosis only so that a potentially morbid surgery has limited therapeutic value. In this study we evaluated the ability of defined serum miRNAs to predict residual viable nonseminomatous germ cell tumors after chemotherapy. MATERIALS AND METHODS: Levels of serum miRNA, including miR-371a-3p, miR-373-3p and miR-367-3p, were measured using the ampTSmiR (amplification targeted serum miRNA) test in 82 patients, including 39 in cohort 1 and 43 in cohort 2, who were treated with orchiectomy, chemotherapy and post-chemotherapy retroperitoneal lymph node dissection. miRNA levels were compared to clinical characteristics and serum tumor markers, and correlated with the presence of viable germ cell tumor vs fibrosis/necrosis and teratoma. ROC analysis was done to determine miRNA discriminative capacity. RESULTS: miRNA levels were significantly associated with disease extent at chemotherapy and they decreased significantly after chemotherapy. Conventional serum tumor marker levels were uninformative after chemotherapy. However, after chemotherapy miRNA levels remained elevated in patients harboring viable germ cell tumor in post-chemotherapy retroperitoneal lymph node dissection specimens. miR-371a-3p demonstrated the highest discriminative capacity for viable germ cell tumors (AUC 0.874, 95% CI 0.774-0.974, p <0.0001). Using an adapted hypothetical cutoff of 3 cm or less for surgical intervention miR-371a-3p correctly stratified all patients with viable residual retroperitoneal germ cell tumors with 100% sensitivity (p = 0.02). CONCLUSIONS: Our study demonstrates for the first time the potential value of miR-371a-3p to predict viable germ cell tumors in residual masses after chemotherapy. Prospective studies are required to confirm clinical usefulness.


Subject(s)
MicroRNAs/blood , Neoplasms, Germ Cell and Embryonal/blood , Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/blood , Testicular Neoplasms/pathology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Cohort Studies , Humans , Lymph Node Excision , Male , Neoplasms, Germ Cell and Embryonal/drug therapy , Orchiectomy , Sensitivity and Specificity , Testicular Neoplasms/drug therapy , Treatment Outcome
3.
Biochem Biophys Res Commun ; 441(4): 911-6, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24216110

ABSTRACT

Breast cancer anti-estrogen resistance 3 (BCAR3) is an SH2-containing signal transducer and is implicated in tumorigenesis of breast cancer cells. In this study, we found that BCAR3 mediates the induction of ERK activation and DNA synthesis by insulin, but not by IGF-1. Specifically, the SH2 domain of BCAR3 is involved in insulin-stimulated DNA synthesis. Differential tyrosine-phosphorylated patterns of the BCAR3 immune complex were detected in insulin and IGF-1 signaling, suggesting that BCAR3 is a distinct target molecule of insulin and IGF-1 signaling. Moreover, microinjection of BCAR3 inhibitory materials inhibited membrane ruffling induced by insulin, while this did not affect insulin-mediated GLUT4 translocation. Taken together, these results demonstrated that BCAR3 plays an important role in the signaling pathways of insulin leading to cell cycle progression and cytoskeleton reorganization, but not GLUT4 translocation.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , DNA Replication/physiology , Glucose Transporter Type 4/metabolism , Insulin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line, Tumor , Guanine Nucleotide Exchange Factors , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/metabolism , Protein Transport , Rats , Signal Transduction , Tyrosine/metabolism
4.
Int J Cancer ; 131(9): 1998-2007, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22328489

ABSTRACT

Previously, we have identified a panel of breast cancer antiestrogen resistance (BCAR) genes. Several of these genes have clinical relevance because mRNA or protein levels associate with tamoxifen resistance or tumor aggressiveness. We postulated that changes in activation status of protein signaling networks induced by BCAR genes may provide better insight into the mechanisms underlying antiestrogen resistance. Key signal transduction pathways were analyzed for changes in activation or expression using reverse-phase protein microarrays probed with 78 antibodies against signaling proteins with known roles in tumorigenesis. We used ZR-75-1-derived cell lines transduced with AKT1, AKT2, BCAR1, BCAR3, BCAR4, EGFR, GRB7, HRAS, HRAS(v12) or HEF1 and MCF7-derived cell lines transduced with BCAR3, BCAR4 or EGFR. In the antiestrogen-resistant cell lines, we observed increased phosphorylation of several pathways involved in cell proliferation and survival. All tamoxifen-resistant cell lines contained high levels of phosphorylated AKT and its biochemically linked substrates Forkhead box O1/3. The activation of ERBB2, ERBB3 and the downstream modulators focal adhesion kinase and SHC were activated in cells with overexpression of BCAR4. Remarkable differences were observed for the levels of activated AMPK alpha1, cyclins, STAT5, STAT6, ERK1/2 and BCL2. The comparison of the cell signaling networks in estrogen-dependent and -independent cell lines revealed biochemically linked kinase-substrate markers that comprised systemically activated signaling pathways involved in tamoxifen resistance. Our results show that this model provides insights into the molecular and cellular mechanisms of breast cancer progression and antiestrogen resistance. This knowledge may help the development of novel targeted treatments.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor Modulators/pharmacology , Protein Interaction Mapping , Signal Transduction , Tamoxifen/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Cell Survival , Crk-Associated Substrate Protein/genetics , Estrogen Receptor alpha/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Shc Signaling Adaptor Proteins/biosynthesis
5.
J Cell Physiol ; 226(7): 1741-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506106

ABSTRACT

Resistance to the antiestrogen tamoxifen remains a major problem in the management of estrogen receptor-positive breast cancer. Knowledge on the resistance mechanisms is needed to develop more effective therapies. Breast cancer antiestrogen resistance 4 (BCAR4) was identified in a functional screen for genes involved in tamoxifen resistance. BCAR4 is expressed in 27% of primary breast tumors. In patients treated with tamoxifen for metastized disease high BCAR4 mRNA levels are associated with reduced clinical benefit and progression-free survival. Regarding tumor aggressiveness high BCAR4 mRNA levels are associated with a shorter metastasis free survival and overall survival. In the present study, we investigated the role of BCAR4 in endocrine resistance. Forced expression of BCAR4 in human ZR-75-1 and MCF7 breast cancer cells resulted in cell proliferation in the absence of estrogen and in the presence of various antiestrogens. Inhibition of estrogen receptor 1 (ESR1) expression with small interfering RNA (siRNA), implied that the BCAR4-induced mechanism of resistance is independent of ESR1. Highly conserved BCAR4 homologues of rhesus monkey, green monkey, and the less conserved common marmoset gene induced tamoxifen-resistant cell proliferation, in contrast to the distant BCAR4 homologues of bovine and rabbit. Injection of BCAR4-expressing ZR-75-1 cells into nude mice resulted in rapidly growing tumors. In silico analysis showed that BCAR4 mRNA is highly expressed in human placenta and oocyte, and absent in other normal tissues. In conclusion, BCAR4 is a strong transforming gene causing estrogen-independent growth and antiestrogen resistance, and induces tumor formation in vivo. Due to its restricted expression, BCAR4 may be a good target for treating antiestrogen-resistant breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Crk-Associated Substrate Protein/metabolism , Drug Resistance, Neoplasm , Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Amino Acid Sequence , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Crk-Associated Substrate Protein/genetics , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , Haplorhini , Humans , Mice , Mice, Nude , Molecular Sequence Data , Oncogenes , Oocytes/metabolism , Placenta/metabolism , Pregnancy , RNA Interference , RNA, Long Noncoding , RNA, Messenger/metabolism , RNA, Untranslated , Rabbits , Time Factors , Transfection , Tumor Burden
6.
Breast Cancer Res Treat ; 120(2): 401-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19412734

ABSTRACT

To unravel the mechanisms underlying failure of endocrine therapy of breast cancer, we have previously executed a functional genetic screen and identified the adaptor protein BCAR1 to be causative for tamoxifen resistance. As a consequence of the manifold of interactions with other proteins, we characterized the contribution of individual protein domains of BCAR1 to anti-estrogen-resistant proliferation of human breast cancer cells. We took advantage of the observation that the closely related family member HEF1 was unable to support long-term anti-estrogen-resistant cell proliferation. Chimerical proteins containing defined domains of BCAR1 and HEF1 were evaluated for anti-estrogen-resistant growth. Exchange of the SH3 and C-terminal domains did not modify the capacity to support cell proliferation. Full support of anti-estrogen resistant proliferation was observed for chimerical molecules containing the central part of BCAR1. The bi-partite SRC-binding site or the Serine-rich domain did not explain the differential capacity of BCAR1. These findings indicate that the differences between BCAR1 and HEF1 with respect to support of anti-estrogen resistance reside in the substrate domain which contains multiple sites for tyrosine phosphorylation. The crucial interactions required for anti-estrogen resistance occur within the substrate domain of BCAR1. Further deciphering of these interactions may resolve the growth regulatory mechanism and provide an explanation for the observation that primary tumors with high levels of BCAR1 are likely to fail on tamoxifen therapy. This information may also help to devise alternative personalized treatment strategies with improved outcome for breast cancer patients.


Subject(s)
Breast Neoplasms/genetics , Crk-Associated Substrate Protein/chemistry , Crk-Associated Substrate Protein/genetics , Drug Resistance, Neoplasm/genetics , Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Estrogen Receptor Modulators/pharmacology , Female , Humans , Phosphoproteins/genetics , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology
7.
Breast Cancer Res Treat ; 114(1): 23-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18351453

ABSTRACT

Endocrine treatment of breast cancer is widely applied and effective. However, in advanced disease cases, the tumors will eventually progress into an estrogen-independent and therapy-resistant phenotype. To elucidate the molecular mechanisms underlying this endocrine therapy failure, we applied retroviral insertion mutagenesis to identify the main genes conferring estrogen independence to human breast cancer cells. Estrogen-dependent ZR-75-1 cells were infected with replication-defective retroviruses followed by selection with the anti-estrogen 4-hydroxy-tamoxifen. In the resulting panel of 79 tamoxifen-resistant cell lines, the viral integrations were mapped within the human genome. Genes located in the immediate proximity of the retroviral integration sites were characterized for altered expression and their capacity to confer anti-estrogen resistance when transfected into breast cancer cells. Out of 15 candidate BCAR (breast cancer anti-estrogen resistance) genes, seven (AKT1, AKT2, BCAR1, BCAR3, EGFR, GRB7, and TRERF1/BCAR2) were shown to directly underlie estrogen independence. Our results show that insertion mutagenesis is a powerful tool to identify BCAR loci, which may provide insights into the molecular and cellular mechanisms of breast tumor progression and therapy resistance thereby offering novel targets for the development of tailor-made therapeutical and prevention strategies.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Estrogens/physiology , Cell Line, Tumor , Estrogens/genetics , Female , Humans , Mutagenesis, Insertional , Retroviridae , Virus Integration
8.
Breast Cancer Res Treat ; 113(2): 253-60, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18299979

ABSTRACT

Purpose Two genes, TSC22 domain family, member 1 (TSC22D1) and prosaposin (PSAP) were identified in an in vitro functional screen for genes having a causative role in tamoxifen resistance. These genes were also present in our previously established 81-gene signature for resistance to first-line tamoxifen therapy. The aim of this study was to investigate the predictive value of these genes for tamoxifen therapy failure in patients with recurrent breast cancer. Experimental Design The mRNA levels of TSC22D1 and PSAP were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) in 223 estrogen receptor-positive primary breast tumors of patients with recurrent disease treated with first-line tamoxifen therapy. The main objective of this study was the length of progression-free survival (PFS). Results High mRNA levels of TSC22D1 and PSAP were significantly associated with shorter PFS and both were independent of the traditional predictive factors (HR = 1.30, 95% CI = 1.04-1.64 P = 0.023; and HR = 1.40, 95% CI = 1.03-1.88, P = 0.029, respectively). In multivariate analysis, patients with high mRNA levels of both genes associated significantly with no clinical benefit (OR = 0.19, 95% CI = 0.06-0.62, P = 0.006) and had the shortest PFS (HR = 2.05, 95% CI = 1.29-3.25, P = 0.002). Conclusion These results confirm our previous in vitro and tumor-related findings and are indicative for the failure of tamoxifen treatment in breast-cancer patients. Both TSC22D1 and PSAP are associated with clinical outcome and may have a functional role in therapy resistance.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Estrogen Receptor Modulators/therapeutic use , Gene Expression Profiling , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Repressor Proteins/genetics , Saposins/genetics , Tamoxifen/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Chemotherapy, Adjuvant , Combined Modality Therapy , Disease-Free Survival , Estrogen Receptor Modulators/pharmacology , Female , Humans , Middle Aged , Prognosis , Radiotherapy, Adjuvant , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology , Treatment Outcome
9.
Cells ; 8(10)2019 10 08.
Article in English | MEDLINE | ID: mdl-31597402

ABSTRACT

BACKGROUND: LDH (lactate dehydrogenase), AFP (alpha-fetoprotein) and ß-HCG (human chorionic gonadotropin) are used in diagnosis and follow-up of testicular germ cell cancer (TGCC) patients. Our aim was to investigate the association between levels of miR-371a-3p, miR-373-3p and miR-367-3p and clinical features in metastatic TGCC. METHODS: relative levels of miR-371a-3p, miR-373-3p and miR-367-3p were evaluated in serum of metastatic TGCC patients. A prospectively included and a retrospectively selected cohort were studied (total patient number = 109). Blood samples were drawn at start of chemotherapy and during follow-up. Serum microRNA (miR) levels were determined using the ampTSmiR test. RESULTS: at start of chemotherapy, miR-371a-3p, miR-373-3p and miR-367-3p levels were positively correlated to LDH. The median level of these miRs was higher in patients who developed a relapse after complete biochemical remission (n = 34) than in those who had complete durable remission (n = 60). Higher levels of miR-367-3p were found in patients with refractory disease (n = 15) compared to those who had complete response. miR levels decreased during the first week of chemotherapy in patients with complete response and stayed below threshold after one year of treatment. CONCLUSION: high miR levels at start of chemotherapy are associated with worse clinical outcome and can assist in early diagnosing of relapses.


Subject(s)
MicroRNAs/blood , Neoplasms, Germ Cell and Embryonal/blood , Testicular Neoplasms/blood , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , Bleomycin/therapeutic use , Chorionic Gonadotropin/blood , Cisplatin/therapeutic use , Cohort Studies , Etoposide/therapeutic use , Humans , L-Lactate Dehydrogenase/blood , Male , Middle Aged , Neoplasms, Germ Cell and Embryonal/drug therapy , Testicular Neoplasms/drug therapy , Young Adult , alpha-Fetoproteins/analysis
10.
Endocr Relat Cancer ; 15(1): 101-11, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18310279

ABSTRACT

Tamoxifen treatment of estrogen-dependent breast cancer ultimately loses its effectiveness due to the development of resistance. From a functional screen for identifying genes responsible for tamoxifen resistance in human ZR-75-1 breast cancer cells, fibroblast growth factor (FGF) 17 was recovered. The aim of this exploratory study was to assess the predictive value of FGF17 and the receptors FGFR1-4 for the type of response to tamoxifen treatment (clinical benefit) and the duration of progression-free survival (PFS) in patients with recurrent breast cancer. mRNA levels of FGF17 and FGFR1-4 were quantified by real-time reverse transcriptase PCR in 285 estrogen receptor-positive breast carcinomas with clinical follow-up. All patients had recurrent disease and were treated with tamoxifen as first-line systemic therapy for local or distant relapse. FGF17 and FGFR1-3 mRNA levels had no significant predictive value for this group of patients. However, high FGFR4 mRNA levels analyzed as a continuous log-transformed variable predicted poor clinical benefit (odds ratio=1.22; P=0.009) and shorter PFS (hazard ratio=1.18; P<0.001). In addition, in multivariable analysis, the predictive value of FGFR4 was independent from the traditional predictive factors. Our analyses show that FGFR4 may play a role in the biological response of the tumor to tamoxifen treatment. In addition, as altered expression of FGF17 causes tamoxifen resistance in vitro, the FGF signaling pathway could be a valuable target in the treatment of breast cancer patients resistant to endocrine treatment.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Neoplasm Recurrence, Local/diagnosis , Receptor, Fibroblast Growth Factor, Type 4/genetics , Tamoxifen/therapeutic use , Adult , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/secondary , Survival Rate , Treatment Failure
11.
Biochem Biophys Res Commun ; 375(3): 430-4, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18722344

ABSTRACT

BCAR3 (breast cancer anti-estrogen resistance 3) is a signal transducer containing an SH2 domain, a proline/serine-rich domain and a GDP-exchange factor homologous domain, whose role in signaling pathways is currently unclear. Furthermore, BCAR3 is implicated in anti-estrogen resistance of breast cancer cells. In the present study, we investigated the functional role of BCAR3 in a mitogenic signaling pathway of EGF in non-tumorigenic human breast epithelial MCF-12A cells. Microinjection of an anti-BCAR3 antibody, siRNAs targeting BCAR3 and an SH2 domain of BCAR3 inhibited EGF-induced DNA synthesis. Direct association of BCAR3 with activated EGF receptor and Cas was observed. Lastly, microinjection of a BCAR3 expression plasmid induced DNA synthesis. These findings suggest that the BCAR3 protein, through its SH2 domain, is involved in the signaling pathways of EGF leading to cell cycle progression, and that BCAR3 itself is part of a mitogenic signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast/cytology , Breast/metabolism , Cell Cycle , DNA/biosynthesis , Epidermal Growth Factor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Antibodies/immunology , Breast/drug effects , Cell Line , DNA/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Guanine Nucleotide Exchange Factors , Humans , Microinjections , Mitosis , RNA, Small Interfering/genetics , src Homology Domains/genetics
12.
Stem Cell Reports ; 11(6): 1493-1505, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30503260

ABSTRACT

Predicting developmental potency and risk of posttransplantation tumor formation by human pluripotent stem cells (hPSCs) and their derivatives largely rely on classical histological analysis of teratomas. Here, we investigated whether an assay based on microRNAs (miRNA) in blood plasma is able to detect potentially malignant elements. Several hPSCs and human malignant germ cell tumor (hGCT) lines were investigated in vitro and in vivo after mouse xenografting. The multiple conventional hPSC lines generated mature teratomas, while xenografts from induced hPSCs (hiPSCs) with reactivated reprogramming transgenes and hGCT lines contained undifferentiated and potentially malignant components. The presence of these elements was reflected in the mRNA and miRNA profiles of the xenografts with OCT3/4 mRNA and the miR-371 and miR-302 families readily detectable. miR-371 family members were also identified in mouse plasma faithfully reporting undifferentiated elements in the xenografts. This study demonstrated that undifferentiated and potentially malignant cells could be detected in vivo.


Subject(s)
Biological Assay/methods , Biomarkers, Tumor/blood , Cell Differentiation/genetics , MicroRNAs/blood , Pluripotent Stem Cells/metabolism , Teratoma/blood , Teratoma/genetics , Animals , Biomarkers, Tumor/genetics , Cell Line , Gene Expression Regulation, Neoplastic , Humans , Mice , MicroRNAs/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Xenograft Model Antitumor Assays
13.
Mol Cancer Res ; 4(6): 379-86, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16778085

ABSTRACT

Antiestrogens, such as tamoxifen, are widely used for endocrine treatment of estrogen receptor-positive breast cancer. However, as breast cancer progresses, development of tamoxifen resistance is inevitable. The mechanisms underlying this resistance are not well understood. To identify genes involved in tamoxifen resistance, we have developed a rapid screening method. To alter the tamoxifen-sensitive phenotype of human ZR-75-1 breast cancer cells into a tamoxifen-resistant phenotype, the cells were infected with retroviral cDNA libraries derived from human placenta, human brain, and mouse embryo. Subsequently, the cells were selected for proliferation in the presence of 4-hydroxy-tamoxifen (OH-TAM) and integrated cDNAs were identified by sequence similarity searches. From 155 OH-TAM-resistant cell colonies, a total of 25 candidate genes were isolated. Seven of these genes were identified in multiple cell colonies and thus cause antiestrogen resistance. The epidermal growth factor receptor, platelet-derived growth factor receptor-alpha, platelet-derived growth factor receptor-beta, colony-stimulating factor 1 receptor, neuregulin1, and fibroblast growth factor 17 that we have identified have been described as key regulators in the mitogen-activated protein kinase pathway. Therefore, this pathway could be a valuable target in the treatment of patients with breast cancer resistant to endocrine treatment. In addition, the putative gene LOC400500, predicted by in silico analysis, was identified. We showed that ectopic expression of this gene, designated as breast cancer antiestrogen resistance 4 (BCAR4), caused OH-TAM resistance and anchorage-independent cell growth in ZR-75-1 cells and that the intact open reading frame was required for its function. We conclude that retroviral transfer of cDNA libraries into human breast cancer cells is an efficient method for identifying genes involved in tamoxifen resistance.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Estrogen Antagonists/pharmacology , Genetic Testing , Tamoxifen/analogs & derivatives , Animals , Brain Chemistry , Cell Division , Cell Line, Tumor , Embryo, Mammalian , Frameshift Mutation , Gene Library , Genomics/methods , Humans , Mice , Placenta/chemistry , Retroviridae/genetics , Tamoxifen/pharmacology , Transduction, Genetic
14.
Oncotarget ; 8(35): 58037-58049, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28938535

ABSTRACT

Multiple studies, including various methods and overall limited numbers of mostly heterogeneous cases, indicate that the level of embryonic stem cell microRNAs (miRs) (e.g. 371a-3p, 372-3p, 373-3p, and 367-3p) are increased in serum at primary diagnosis of almost all testicular germ cell cancer (TGCC). Here we determine the status of three of these miRs in serum samples of 250 TGCC patients, collected at time of primary diagnosis, compared with 60 non-TGCC patients and 104 male healthy donors. The levels of miRs were measured by the robust ampTSmiR test, including magnetic bead-based miR isolation and target specific pre-amplification followed by real-time quantitative PCR (RT-qPCR) detection. Calibration is performed based on the non-human spike-in ath-miR-159a, and normalization on the endogenous control miR-30b-5p. The serum levels of miR-371a-3p, 373-3p, and 367-3p are informative to accurately detect TGCC patients, both seminomas and non-seminomas, at the time of primary diagnosis (p< 0.000). Receiver Operating Characteristic (ROC) analysis demonstrate that the Area Under the Curve (AUC) for miR-371a-3p is 0.951 (being 0.888 for miR-373-3p and 0.861 for miR-367-3p), with a sensitivity of 90%, and a specificity of 86% (positive predictive value of 94% and negative predictive value of 79%). Inclusion of miR-373-3p and 367-3p resulted in a AUC of 0.962, with a 90% sensitivity and 91% specificity. Similar results were obtained using the raw Ct data. Importantly, the results demonstrate that ampTSmiR is not suitable to detect pure teratoma as well as the precursor of TGCC, i.e., Germ Cell Neoplasia In Situ (GCNIS). The largest series evaluated so far, demonstrate that detection of the embryonic stem cell miR-371a-3p, 373-3p and 367-3p is highly informative to diagnose patients with a primary TGCC.

15.
Cell Oncol (Dordr) ; 40(4): 379-388, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28612337

ABSTRACT

PURPOSE: α-fetoprotein (AFP) and human chorionic gonadotropin subunit beta (B-HCG) are informative serum biomarkers for the primary diagnosis and follow-up of testicular germ cell cancer (TGCC) patients. About 20% of TGCC patients with a non-seminoma (NS) and about 80% with a seminoma (SE) are, however, negative for these biomarkers. Embryonic stem cell microRNAs (miRs) may serve as promising alternative serum biomarkers. Here we investigated a retrospective series of serum samples from selected TGCC patients who developed a relapse in time to test the possible additional value of the serum-based ampTSmiR test compared to the conventional serum-based protein biomarkers for follow-up. METHODS: We investigated 261 retrospective serum samples of six selected fully evaluated TGCC patients with a proven relapse using the ampTSmiR test for miR-371a-3p, miR-373-3p, and miR-367-3p and compared the results to those of the conventional protein biomarkers. RESULTS: At primary diagnosis, elevated serum B-HCG, AFP and LDH levels were found to be informative in 4/6, 3/6 and 3/6 patients, respectively. At primary diagnosis the levels of miR-371a-3p and miR-373-3p were elevated in 4/4, and miR-367-3p in 3/4 patients. For two cases no starting serum sample was available for retrospective miR analysis. Residual disease (overlooked by histopathological examination) was detected in one case by miR-371a-3p only. The miR-371a-3p level was increased in one patient two months before detection of an intracranial metastasis. B-HCG was informative in 3/4 and the ampTSmiR test in 4/4 patients with a relapse or residual disease. None of the biomarkers were informative for the detection of residual mature teratoma. CONCLUSIONS: The ampTSmiR test is more sensitive than the conventional TGCC protein biomarkers for the detection of residual disease and relapse, excluding mature teratoma.


Subject(s)
Biomarkers, Tumor/genetics , MicroRNAs/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Testicular Neoplasms/genetics , Adult , Biomarkers, Tumor/blood , Humans , Male , MicroRNAs/blood , Neoplasm Recurrence, Local , Neoplasms, Germ Cell and Embryonal/blood , Neoplasms, Germ Cell and Embryonal/diagnosis , Prognosis , Retrospective Studies , Testicular Neoplasms/blood , Testicular Neoplasms/diagnosis , Young Adult
16.
Breast Cancer Res ; 7(1): R82-92, 2005.
Article in English | MEDLINE | ID: mdl-15642172

ABSTRACT

INTRODUCTION: Tamoxifen is effective for endocrine treatment of oestrogen receptor-positive breast cancers but ultimately fails due to the development of resistance. A functional screen in human breast cancer cells identified two BCAR genes causing oestrogen-independent proliferation. The BCAR1 and BCAR3 genes both encode components of intracellular signal transduction, but their direct effect on breast cancer cell proliferation is not known. The aim of this study was to investigate the growth control mediated by these BCAR genes by gene expression profiling. METHODS: We have measured the expression changes induced by overexpression of the BCAR1 or BCAR3 gene in ZR-75-1 cells and have made direct comparisons with the expression changes after cell stimulation with oestrogen or epidermal growth factor (EGF). A comparison with published gene expression data of cell models and breast tumours is made. RESULTS: Relatively few changes in gene expression were detected in the BCAR-transfected cells, in comparison with the extensive and distinct differences in gene expression induced by oestrogen or EGF. Both BCAR1 and BCAR3 regulate discrete sets of genes in these ZR-75-1-derived cells, indicating that the proliferation signalling proceeds along distinct pathways. Oestrogen-regulated genes in our cell model showed general concordance with reported data of cell models and gene expression association with oestrogen receptor status of breast tumours. CONCLUSIONS: The direct comparison of the expression profiles of BCAR transfectants and oestrogen or EGF-stimulated cells strongly suggests that anti-oestrogen-resistant cell proliferation is not caused by alternative activation of the oestrogen receptor or by the epidermal growth factor receptor signalling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Genes, BRCA1 , Receptors, Estrogen/physiology , Adaptor Proteins, Signal Transducing/genetics , Antineoplastic Agents, Hormonal/pharmacology , BRCA1 Protein/biosynthesis , Drug Resistance, Neoplasm/genetics , ErbB Receptors/physiology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors , Humans , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tamoxifen/pharmacology , Transfection , Tumor Cells, Cultured
17.
PLoS One ; 10(8): e0136845, 2015.
Article in English | MEDLINE | ID: mdl-26317614

ABSTRACT

BACKGROUND: Most breast cancers depend on estrogenic growth stimulation. Functional genetic screenings in in vitro cell models have identified genes, which override growth suppression induced by anti-estrogenic drugs like tamoxifen. Using that approach, we have previously identified Breast Cancer Anti-Estrogen Resistance 4 (BCAR4) as a mediator of cell proliferation and tamoxifen-resistance. Here, we show high level of expression and function of BCAR4 in human breast cancer. METHODS: BCAR4 mRNA expression was evaluated by (q)RT-PCR in a panel of human normal tissues, primary breast cancers and cell lines. A new antibody raised against C78-I97 of the putative BCAR4 protein and used for western blot and immunoprecipitation assays. Furthermore, siRNA-mediated gene silencing was implemented to study the function of BCAR4 and its downstream targets ERBB2/3. RESULTS: Except for placenta, all human normal tissues tested were BCAR4-negative. In primary breast cancers, BCAR4 expression was comparatively rare (10%), but associated with enhanced proliferation. Relative high BCAR4 mRNA expression was identified in IPH-926, a cell line derived from an endocrine-resistant lobular breast cancer. Moderate BCAR4 expression was evident in MDA-MB-134 and MDA-MB-453 breast cancer cells. BCAR4 protein was detected in breast cancer cells with ectopic (ZR-75-1-BCAR4) and endogenous (IPH-926, MDA-MB-453) BCAR4 mRNA expression. Knockdown of BCAR4 inhibited cell proliferation. A similar effect was observed upon knockdown of ERBB2/3 and exposure to lapatinib, implying that BCAR4 acts in an ERBB2/3-dependent manner. CONCLUSION: BCAR4 encodes a functional protein, which drives proliferation of endocrine-resistant breast cancer cells. Lapatinib, a clinically approved EGFR/ERBB2 inhibitor, counteracts BCAR4-driven tumor cell growth, a clinical relevant observation.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Lobular/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Adult , Aged , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Lapatinib , Middle Aged , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism
18.
Int J Dev Biol ; 57(2-4): 241-53, 2013.
Article in English | MEDLINE | ID: mdl-23784835

ABSTRACT

Human germ cell tumors are of interest because of their epidemiology, clinic and patho-biology. Histologically, they are subdivided into various elements, with similarities to embryogenesis. Recent insight triggered development of a higher order division into five types of human germ cell tumors. In the context of male germ cells, only three are relevant; Type I: teratomas and yolk sac tumors of neonates and infants; Type II: seminomas and nonseminomas of (predominantly) adolescents and adults; and Type III: spermatocytic seminomas of the elderly. Various animal models, both occurring spontaneous or induced, are reported, of which their relevance is still a matter of debate. Recent multidisciplinary studies have led to a significant increase in our understanding of the parameters involved in the earliest pathogenetic steps of human germ cells tumors, particularly the seminomas and nonseminomas (Type II). This paper will discuss a number of interesting insights into the normal and aberrant regulation of germ cell development, resulting in the so-called genvironmental hypothesis. This assumes a subtle interaction between environmental- and (epi)genetic parameters, resulting in clinical/phenotypical characteristics. These influence signaling pathways and thereby developmental processes, including gonadal development, germ cell proliferation, maturation and apoptosis. In the case of a disturbed physiology, either due to the germ cell itself, or the micro-environment, embryonic germ cells, during a specific window of sensitization, might be blocked in their maturation, resulting in carcinoma in situ or gonadoblastoma, the precursors of seminomas and nonseminomas. The level of testicularization of the gonad determines the histological composition of the precursor. These insights will allow a better definition of individuals at risk of developing a germ cell malignancy, and allow a better selection of scientific approaches to elucidate the corresponding pathogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Gene-Environment Interaction , Germ Cells/cytology , Neoplasms, Germ Cell and Embryonal/etiology , Signal Transduction , Testicular Neoplasms/etiology , Animals , Disease Models, Animal , Germ Cells/physiology , Humans , Male , Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/pathology
19.
Endocr Relat Cancer ; 17(1): 215-30, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19966015

ABSTRACT

Although endocrine treatment of breast cancer is effective and common practice, in advanced disease the development of resistance is nearly inevitable. To get more insight into individual genes that account for resistance against hormonal agents, we have executed functional genetic screens and subsequently evaluated the clinical relevance of several identified genes with respect to tumor aggressiveness and tamoxifen resistance in estrogen receptor-positive patients. Estrogen-dependent human breast cancer cells were transduced with different retroviral cDNA expression libraries and subjected to selective cultures with various anti-estrogens. From a total of 264 resistant cell clones, 132 different genes were recovered by PCR. By applying stringent selection criteria, we identified 15 breast cancer anti-estrogen resistance (BCAR) genes individually yielding resistance. BCAR genes were recovered with differential frequencies for the diverse culture conditions and anti-estrogen drugs. Analysis of the relation of BCAR genes (EIF1, FBXL10, HRAS, NRG1, PDGFRA, PDGFRB, RAD21, and RAF1) with tamoxifen treatment in patients with advanced disease showed significant association with clinical benefit and progression-free survival for EIF1 and PDGFRA mRNA levels. Furthermore, PDGFRA and HRAS mRNA levels were significantly associated with tumor aggressiveness in lymph node-negative patients who had not received adjuvant systemic therapy. In conclusion, our functional genetic screens showed that BCAR genes differ in their ability to confer resistance towards distinct anti-estrogens. Based on the clinical relevance of several BCAR genes, further studies are warranted to characterize the underlying mechanisms, which may ultimately lead to the development of novel treatments and more individualized management of breast cancer patients.


Subject(s)
Biomarkers, Pharmacological , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Estrogen Antagonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Tamoxifen/therapeutic use , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents, Hormonal/pharmacology , Biomarkers, Pharmacological/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Crk-Associated Substrate Protein/genetics , Crk-Associated Substrate Protein/metabolism , Disease Progression , Drug Resistance, Neoplasm/drug effects , Estrogen Antagonists/metabolism , Female , Genetic Testing , Guanine Nucleotide Exchange Factors , HeLa Cells , Humans , Mice , Middle Aged , Neoplasm Invasiveness , Retrospective Studies , Substrate Specificity
20.
J Clin Oncol ; 27(4): 542-9, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19075277

ABSTRACT

PURPOSE: We have previously identified a set of breast cancer antiestrogen resistance (BCAR) genes causing estrogen independence and tamoxifen resistance in vitro using a functional genetic screen. Here, we explored whether these BCAR genes provide predictive value for tamoxifen resistance and prognostic information for tumor aggressiveness in breast cancer patients. PATIENTS AND METHODS: mRNA levels of 10 BCAR genes (AKT1, AKT2, BCAR1, BCAR3, EGFR, ERBB2, GRB7, SRC, TLE3, and TRERF1) were measured in estrogen receptor-positive breast tumors using quantitative reverse-transcriptase polymerase chain reaction. Normalized mRNA levels were evaluated for association with progression-free survival (PFS) in 242 patients receiving tamoxifen as first-line monotherapy for recurrent disease, and with distant metastasis-free survival (MFS) in 413 lymph node-negative (LNN) primary breast cancer patients who did not receive systemic adjuvant therapy. RESULTS: Concerning tamoxifen resistance, BCAR3, ERBB2, GRB7, and TLE3 mRNA levels were predictive for PFS, independent of traditional predictive factors. By combining GRB7 (or ERBB2) and TLE3 mRNA levels, patients could be classified in three subgroups with distinct PFS. For the evaluation of tumor aggressiveness, AKT2, EGFR, and TRERF1 mRNA levels were all significantly associated with MFS, independent of traditional prognostic factors. Using the combined AKT2 and EGFR mRNA status, four prognostic groups were identified with different MFS outcomes. CONCLUSION: The majority of BCAR genes, which were revealed to confer tamoxifen resistance and estrogen independence in vitro by functional screening, have clinical relevance, and associate with tamoxifen resistance and/or tumor aggressiveness in breast cancer patients.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Tamoxifen/therapeutic use , Adult , Aged , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Disease Progression , Female , Humans , Middle Aged , RNA, Messenger/analysis , Receptors, Estrogen/analysis , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL