Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Zdr Varst ; 60(4): 199-209, 2021 Dec.
Article En | MEDLINE | ID: mdl-34917188

AIM: With the aim of improving personalized treatment of patients on chemotherapy, the objective of the study was to assess the degree of association between selected Quality of life (QoL) indicators and both clinical and imaging cardiac status indicators when detecting deterioration in QoL of these patients. METHODS: In a cohort clinical study in Hamburg, from August 2017 through October 2020, 59 cancer patients, aged 18-80 years, were evaluated before chemotherapy, and at several follow-ups, using EQ-5D and SF-36 QoL questionnaires, fast strain-encoded (fast-SENC) cardiac magnetic resonance (CMR), conventional CMR, and echocardiography, and further received a clinical and biomarker examination. Data was analyzed using survival analyses. A decline of more than 5% in each observed QoL metric value was defined as the observed event. Patient were separated into groups according to the presentation of cardiotoxicity as per its clinical definition, the establishment of the indication for cardioprotective therapy initiation, and by a worsening in the value of each observed imaging metric by more than 5% in the previous follow-up compared to the corresponding pre-chemotherapy baseline value. RESULTS: Among clinical cardiac status indicators, the indication for cardioprotective therapy showed statistically good association with QoL scores (EQ-5D p=0.028; SF-36 physical component p=0.016; SF-36 mental component p=0.012). In terms of imaging metrics, the MyoHealth segmental myocardial strain score was the only one demonstrating consistently good QoL score association (EQ-5D p=0.005; SF-36 physical component p=0.056; SF-36 mental component p=0.002). CONCLUSIONS: Established fast-SENC CMR scores are capable of highlighting patients with reduced QoL, who require more frequent/optimal management.

2.
Circ Cardiovasc Imaging ; 14(6): e012459, 2021 06.
Article En | MEDLINE | ID: mdl-34126756

BACKGROUND: Our goal was to evaluate the ability of cardiovascular magnetic resonance for detecting and predicting cardiac dysfunction in patients receiving cancer therapy. Left ventricular ejection fraction, global and regional strain utilizing fast-strain-encoded, T1 and T2 mapping, and cardiac biomarkers (troponin and BNP [brain natriuretic peptide]) were analyzed. METHODS: Sixty-one patients (47 with breast cancer, 11 with non-Hodgkin lymphoma, and 3 with Hodgkin lymphoma) underwent cardiovascular magnetic resonance scans at baseline and at regular intervals during 2 years of follow-up. The percentage of all left ventricular myocardial segments with strain ≤-17% (normal myocardium [%]) was analyzed. Clinical cardiotoxicity (CTX) and sub-CTX were defined according to standard measures. RESULTS: Nine (15%) patients developed CTX, 26 (43%) had sub-CTX. Of the 35 patients with CTX or sub-CTX, 24 (69%) were treated with cardioprotective medications and showed recovery of cardiac function. The amount of normal myocardium (%) exhibited markedly higher accuracy for the detection of CTX and sub-CTX compared with left ventricular ejection fraction, T1, and T2 mapping as well as troponin I (Δareas under the curve=0.20, 0.24, and 0.46 for normal myocardium (%) versus left ventricular ejection fraction, troponin I, and T1 mapping, P<0.001 for all). In addition, normal myocardium (%) at baseline accurately identified patients with subsequent CTX (P<0.001), which was not achieved by any other markers. CONCLUSIONS: Normal myocardium (%) derived by fast-strain-encoded cardiovascular magnetic resonance, is an accurate and sensitive tool that can establish cardiac safety in patients with cancer undergoing cardiotoxic chemotherapy not only for the early detection but also for the prediction of those at risk of developing CTX. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03543228.


Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Early Diagnosis , Heart Diseases/diagnosis , Magnetic Resonance Imaging, Cine/methods , Natriuretic Peptide, Brain/blood , Troponin I/blood , Aged , Biomarkers/blood , Breast Neoplasms/blood , Cardiotoxicity , Female , Follow-Up Studies , Heart Diseases/blood , Heart Diseases/physiopathology , Humans , Male , Middle Aged , Myocardial Contraction/physiology , Predictive Value of Tests , Prospective Studies , Time Factors , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
3.
Future Oncol ; 16(14): 939-953, 2020 05.
Article En | MEDLINE | ID: mdl-32298187

Aim: To determine quality of life, effectiveness and safety of oral netupitant-palonosetron (NEPA)-based antiemetic prophylaxis in the real-world setting. Materials & methods: Prospective, noninterventional study in adults receiving highly or moderately emetogenic chemotherapy and NEPA for three cycles. NEPA was administered per summary of product characteristics. Results: A total of 2429 patients enrolled, 2173 were evaluable. 'No impact on daily life' due to vomiting was reported by 85%/82% of patients in the highly emetogenic chemotherapy/moderately emetogenic chemotherapy groups in cycle 1, with rates of 54%/59% for nausea. Overall, complete response rate was 89%/87%/83% in the acute/delayed/overall phases. NEPA was well tolerated. Conclusion: NEPA had beneficial effects on the quality of life of a heterogeneous group of cancer patients and was safe and effective in the real-world setting.


Antineoplastic Combined Chemotherapy Protocols/adverse effects , Nausea/etiology , Nausea/prevention & control , Palonosetron/therapeutic use , Pyridines/therapeutic use , Vomiting/etiology , Vomiting/prevention & control , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Neoplasms/complications , Neoplasms/drug therapy , Palonosetron/administration & dosage , Palonosetron/adverse effects , Prospective Studies , Pyridines/administration & dosage , Pyridines/adverse effects , Quality of Life , Treatment Outcome
4.
N Engl J Med ; 380(7): 617-628, 2019 02 14.
Article En | MEDLINE | ID: mdl-30516102

BACKGROUND: Patients who have residual invasive breast cancer after receiving neoadjuvant chemotherapy plus human epidermal growth factor receptor 2 (HER2)-targeted therapy have a worse prognosis than those who have no residual cancer. Trastuzumab emtansine (T-DM1), an antibody-drug conjugate of trastuzumab and the cytotoxic agent emtansine (DM1), a maytansine derivative and microtubule inhibitor, provides benefit in patients with metastatic breast cancer that was previously treated with chemotherapy plus HER2-targeted therapy. METHODS: We conducted a phase 3, open-label trial involving patients with HER2-positive early breast cancer who were found to have residual invasive disease in the breast or axilla at surgery after receiving neoadjuvant therapy containing a taxane (with or without anthracycline) and trastuzumab. Patients were randomly assigned to receive adjuvant T-DM1 or trastuzumab for 14 cycles. The primary end point was invasive disease-free survival (defined as freedom from ipsilateral invasive breast tumor recurrence, ipsilateral locoregional invasive breast cancer recurrence, contralateral invasive breast cancer, distant recurrence, or death from any cause). RESULTS: At the interim analysis, among 1486 randomly assigned patients (743 in the T-DM1 group and 743 in the trastuzumab group), invasive disease or death had occurred in 91 patients in the T-DM1 group (12.2%) and 165 patients in the trastuzumab group (22.2%). The estimated percentage of patients who were free of invasive disease at 3 years was 88.3% in the T-DM1 group and 77.0% in the trastuzumab group. Invasive disease-free survival was significantly higher in the T-DM1 group than in the trastuzumab group (hazard ratio for invasive disease or death, 0.50; 95% confidence interval, 0.39 to 0.64; P<0.001). Distant recurrence as the first invasive-disease event occurred in 10.5% of patients in the T-DM1 group and 15.9% of those in the trastuzumab group. The safety data were consistent with the known safety profile of T-DM1, with more adverse events associated with T-DM1 than with trastuzumab alone. CONCLUSIONS: Among patients with HER2-positive early breast cancer who had residual invasive disease after completion of neoadjuvant therapy, the risk of recurrence of invasive breast cancer or death was 50% lower with adjuvant T-DM1 than with trastuzumab alone. (Funded by F. Hoffmann-La Roche/Genentech; KATHERINE ClinicalTrials.gov number, NCT01772472 .).


Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/drug therapy , Maytansine/analogs & derivatives , Trastuzumab/therapeutic use , Ado-Trastuzumab Emtansine , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/adverse effects , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Lymphatic Metastasis , Maytansine/adverse effects , Maytansine/therapeutic use , Middle Aged , Neoadjuvant Therapy , Neoplasm Metastasis , Neoplasm Staging , Neoplasm, Residual , Peripheral Nervous System Diseases/chemically induced , Radiotherapy , Receptor, ErbB-2/metabolism , Trastuzumab/adverse effects , Treatment Outcome , Young Adult
5.
Arch Gynecol Obstet ; 296(6): 1153-1160, 2017 Dec.
Article En | MEDLINE | ID: mdl-28980126

PURPOSE: The protein tyrosine phosphatase PRL-3 plays an important role in cancer cell migration, invasion and metastasis. In breast cancer, PRL-3 is overexpressed in 70-75% of tumors and even more frequently in lymph node metastases. Moreover, PRL-3 overexpression in breast cancer is associated with an adverse disease outcome. Aim of this study was to determine the role of PRL-3 in breast cancer cell proliferation, migration and invasion in vitro. METHODS: PRL-3 mRNA expression was evaluated in 6 breast cancer cell lines by quantitative real-time PCR. To investigate the effect of PRL-3 expression in breast cancer cells in vitro we both up- and downregulated PRL-3 expression in breast cancer cells and performed in vitro wound repair cell motility assays and invasion assays. The influence of PRL-3 knockdown in MCF-7 cells on the expression of several gene products involved in cell invasion and cytoskeletal function was evaluated with real-time PCR. RESULTS: PRL-3 mRNA expression was demonstrated in all breast cancer cell lines evaluated. Knockdown of PRL-3 in MCF-7 cells resulted in decreased proliferation, wound healing and invasion. PRL-3 knockdown in MCF-7 cells resulted in a significant reduction of heparanase, MMP-9, actin gamma-2 and Myosin 9 expression, and significant elevation of E-cadherin. CONCLUSIONS: We conclude that PRL-3 is an important regulatory factor for breast cancer cell proliferation and invasion. Loss of PRL-3 function induces an antimetastatic gene expression profile in breast cancer cells. Due to its role in tumor growth and metastasis, PRL-3 emerges as a new therapeutic target in breast cancer therapy.


Breast Neoplasms/genetics , Cell Movement/genetics , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/genetics , Antigens, CD , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cytoskeleton , Female , Gene Expression , Gene Expression Regulation, Neoplastic/genetics , Humans , Lymphatic Metastasis , MCF-7 Cells , Matrix Metalloproteinase 9/genetics , Neoplasm Invasiveness/pathology , Real-Time Polymerase Chain Reaction
6.
Gynecol Oncol ; 117(1): 134-8, 2010 Apr.
Article En | MEDLINE | ID: mdl-20071013

OBJECTIVE: Adiponectin serum levels have been shown to be inversely correlated with breast cancer risk. The protein is believed to act through adiponectin receptor 1 (AdipoR1) and has been suggested to play an important role in cancer development. While AdipoR1 is known to be expressed in invasive tumors, its role in DCIS remains elusive. We therefore investigated AdipoR1 expression in both invasive and preinvasive breast cancer. METHODS: Tissue microarrays were established from paraffin-embedded archived tissues which contained 104 invasive breast cancers with adjacent preinvasive component (DCIS) as well as 96 preinvasive breast cancers. AdipoR1 expression was investigated by immunohistochemistry and correlated with clinical and tumor parameters. RESULTS: AdipoR1 was detected in stromal and epithelial components of both invasive and preinvasive breast cancer. However, stromal and epithelial immunoreactivity for AdipoR1 was significantly higher in invasive breast cancer compared to preinvasive DCIS (p<0.001 and p=0.009). Within DCIS, AdipoR1 expression was inversely correlated with tumor size (r=-0.238, p=0.033). Menopausal status showed no influence on AdipoR1 expression. CONCLUSIONS: The altered expression of AdipoR1 in invasive breast cancer compared to DCIS suggests that the receptor-binding protein adiponectin might exert growth inhibitory effects that are overcome in transformation of preinvasive to invasive breast cancer.


Breast Neoplasms/metabolism , Carcinoma in Situ/metabolism , Carcinoma, Ductal, Breast/metabolism , Receptors, Adiponectin/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/pathology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Humans , Immunohistochemistry , Microarray Analysis , Middle Aged , Neoplasm Invasiveness
7.
Int J Cancer ; 127(3): 696-706, 2010 Aug 01.
Article En | MEDLINE | ID: mdl-19960438

Human epidermal growth factor receptor 2 (HER2) is frequently overexpressed in human breast cancers. It is known to drive tumor growth and progression and represents a prominent target in breast cancer therapy. The endothelin (ET) system, in particular ET-1 and its receptor ET(A)R, is of major relevance for breast cancer growth and invasion. Having previously demonstrated coexpression of ET(A)R and HER2 in breast tumors, this study was designed to investigate molecular interactions of HER2 (including the epidermal growth factor receptor EGFR as its major coreceptor) and ET signaling, and the potential benefit of a combined anti-HER2/ET(A)R treatment in human breast cancer cells. Dual HER2-ET(A)R targeting utilizing trastuzumab (monoclonal anti-HER2 antibody) and the ET(A)R antagonist atrasentan was superior to each agent alone in inhibiting basal and EGF-induced proliferation and invasion of HER2-overexpressing BT-474 and SK-BR-3 cells. EGF-induced invasion was partially inhibited by atrasentan alone, suggesting the involvement of ET(A)R in EGF receptor mediated invasion of breast cancer cells. Moreover, secretion of the pro-invasive ET-1 was shown to be induced by EGF via EGFR and HER2, including MAPK-dependent signaling. In turn, an ET-1/ET(A)R-dependent regulation of EGFR protein expression and phosphorylation (at Tyr845) was observed, which may contribute to the additional anti-proliferative and anti-invasive effects of atrasentan on trastuzumab treated cells; reconfirming, atrasentan failed to enhance inhibitory effects of EGFR-targeted agents. This study suggests complex interactions between HER2/EGFR and ET pathways in breast cancer and supports the hypothesis that dual HER2-ET(A)R targeting may represent a highly effective approach in breast cancer treatment.


Antibodies, Monoclonal/pharmacology , Breast Neoplasms/pathology , Genes, erbB-2 , Neoplasm Metastasis/prevention & control , Receptor, Endothelin A/drug effects , Antibodies, Monoclonal, Humanized , Atrasentan , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Down-Regulation , Drug Synergism , Endothelin-1/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Phosphorylation , Pyrrolidines/pharmacology , Receptor, Endothelin A/metabolism , Trastuzumab
8.
Breast Cancer Res Treat ; 123(2): 345-57, 2010 Sep.
Article En | MEDLINE | ID: mdl-19943105

Endothelin-1 (ET-1) and endothelin A receptor (ETAR) contribute to the development and progression of breast carcinomas by modulating cell proliferation, angiogenesis, and anti-apoptosis. We investigated antitumoral effects of the specific ETAR antagonist ZD4054 in breast cancer cells and xenografts, and assessed antitumoral efficacy of the combinations of ZD4054 with aromatase inhibitors and fulvestrant. Gene expression changes were assessed by quantitative real-time PCR. Cell proliferation was measured using alamarBlue; migration and invasion assays were performed using modified Boyden chambers. Evaluating the antitumoral efficacy of ZD4054 in vivo, different breast cancer models were employed using nude mice xenografts. ZD4054 reduced ET-1 and ETAR expression in MCF-7, MDA-MB-231, and MDA-MB-468 breast cancer cells in a concentration-dependent manner. ZD4054 inhibited invasion by up to 37.1% (P = 0.022). Combinations of ZD4054 with either anastrozole or letrozole produced significant reductions in migration of aromatase-overexpressing MCF-7aro cells (P < 0.05). Combination of ZD4054 with fulvestrant reduced MCF-7 cell migration and invasion by 36.0% (P = 0.027) and 56.7% (P < 0.001), respectively, with effects significantly exceeding those seen with either compound alone. Regarding tumor volume reduction in vivo, ZD4054 (10 mg/kg) was equipotent to fulvestrant (200 mg/kg) and exhibited additive effects with anastrozole (0.5 mg/kg). These data are the first indicating that selective ETAR antagonism by ZD4054 displays antitumoral activity on breast cancer cells in vitro and in vivo. Our data strongly support a rationale for the clinical use of ZD4054 in combination with endocrine therapies.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Endothelin A Receptor Antagonists , Anastrozole , Animals , Aromatase Inhibitors/administration & dosage , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endothelin-1/metabolism , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estrogen Antagonists/administration & dosage , Female , Fulvestrant , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Nitriles/administration & dosage , Pyrrolidines/administration & dosage , Receptor, Endothelin A/metabolism , Tamoxifen/administration & dosage , Time Factors , Triazoles/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Anticancer Res ; 29(6): 2167-71, 2009 Jun.
Article En | MEDLINE | ID: mdl-19528477

BACKGROUND: Estrogen receptors (ER) alpha and beta play an important role in breast cancer. Recently, systemic adjuvant endocrine therapy with selective estrogen receptor modulator (SERM) tamoxifen has been challenged by aromatase inhibitors. Compared to antiestrogens, third-generation aromatase inhibitors (anastrozole and letrozole) exhibit an improved efficacy and tolerability. MATERIALS AND METHODS: Using real-time PCR analysis, 21 breast cancer tissue samples were analysed for a change of the ERalpha/ERbeta ratio during malignant progression. In stimulation experiments, differential effects of SERMs, ER antagonists and aromatase inhibitors have been investigated. RESULTS: Transition from normal breast to grade 1 tumors was characterized by down-regulation of ERbeta (relative quantification [RQ]=0.83, p=0.019), while transition from grade 1 to grade 3 tumors was associated with the decrease of ERalpha expression (RQ=1.14 vs. RQ=0.65, p<0.001). In stimulation assays, tamoxifen and fulvestrant increased ERalpha expression to RQ=1.51 (p=0.01) and RQ=1.42 (p<0.001), respectively, and left ERbeta unchanged. In contrast, aromatase inhibitors up-regulated ERbeta to RQ=1.23 (anastrozole, p=0.029) and RQ=1.38 (letrozole, p=0.048). CONCLUSION: Taken together, data indicate that SERMs/antiestrogens and aromatase inhibitors exhibit opposed effects on the ER expression of breast cancer cells: tamoxifen and fulvestrant up-regulate ERalpha expression, while aromatase inhibitors increase ERbeta expression, which may contribute to the aromatase inhibitors' therapeutic superiority over antiestrogens.


Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Anastrozole , Blotting, Western , Breast Neoplasms/metabolism , Estradiol/analogs & derivatives , Estradiol/therapeutic use , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Female , Fulvestrant , Humans , Letrozole , Neoplasm Staging , Neoplasms, Hormone-Dependent/metabolism , Nitriles/therapeutic use , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/therapeutic use , Triazoles/therapeutic use , Tumor Cells, Cultured
10.
Carcinogenesis ; 30(3): 397-407, 2009 Mar.
Article En | MEDLINE | ID: mdl-19126645

The heparan sulfate proteoglycan syndecan-1 (Sdc1) modulates cell proliferation, adhesion, migration and angiogenesis. Proteinase-mediated shedding converts Sdc1 from a membrane-bound coreceptor into a soluble effector capable of binding the same ligands. In breast carcinomas, Sdc1 overexpression correlates with poor prognosis and an aggressive phenotype. To distinguish between the roles of membrane-bound and shed forms of Sdc1 in breast cancer progression, human MCF-7 breast cancer cells were stably transfected with plasmids overexpressing wild-type (WT), constitutively shed and uncleavable forms of Sdc1. Overexpression of WT Sdc1 increased cell proliferation, whereas overexpression of constitutively shed Sdc1 decreased proliferation. Fibroblast growth factor-2-mediated mitogen-activated protein kinase signaling was reduced following small-interfering RNA (siRNA)-mediated knockdown of Sdc1 expression. Constitutively, membrane-bound Sdc1 inhibited invasiveness, whereas soluble Sdc1 promoted invasion of MCF-7 cells into matrigel matrices. The latter effect was reversed by the matrix metalloproteinase inhibitors N-isobutyl-N-(4-methoxyphenylsufonyl) glycyl hydroxamic acid and tissue inhibitor of metalloproteinase (TIMP)-1. Affymetrix microarray analysis identified TIMP-1, Furin and urokinase-type plasminogen activator receptor as genes differentially regulated in soluble Sdc1-overexpressing cells. Endogenous TIMP-1 expression was reduced in cells overexpressing soluble Sdc1 and increased in those overexpressing the constitutively membrane-bound Sdc1. Moreover, E-cadherin protein expression was downregulated in cells overexpressing soluble Sdc1. Our results suggest that the soluble and membrane-bound forms of Sdc1 play different roles at different stages of breast cancer progression. Proteolytic conversion of Sdc1 from a membrane-bound into a soluble molecule marks a switch from a proliferative to an invasive phenotype, with implications for breast cancer diagnostics and potential glycosaminoglycan-based therapies.


Breast Neoplasms/metabolism , Cell Membrane/metabolism , Cell Proliferation , Syndecan-1/physiology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Hydroxamic Acids/pharmacology , MAP Kinase Signaling System/physiology , Mutation , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Sulfonamides/pharmacology , Syndecan-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
11.
Expert Rev Anticancer Ther ; 8(9): 1481-93, 2008 Sep.
Article En | MEDLINE | ID: mdl-18759699

The endothelin system comprises the three peptide hormones endothelin (ET)-1, -2, -3, their G protein-coupled receptors, endothelin-A-receptor (ET(A)R) and endothelin-B-receptor (ET(B)R), and the enzymes of endothelin biosynthesis and degradation. In the past two decades, an impressive amount of data has been accumulated investigating the role of the endothelin system in a variety of malignancies. In many cancers, ET-1/ET(A)R interaction induces proliferation, angiogenesis, antiapoptosis and resistance to chemotherapy. Data indicate a pivotal role of the endothelin system in tumorigenesis, local progression and metastasis. Subsequently, novel drugs have been designed inhibiting ET-1 biosynthesis or ET(A)R interaction. A wide range of preclinical data is available on the role of ET(A)R antagonists in gynecological, urological and breast cancers providing evidence for their antiangiogenic, proapoptotic and growth inhibitory effects. Of particular interest is the anti-invasive and antimetastatic efficacy of ET(A)R antagonists and synergism when co-administered with established cancer therapies. Data indicate a future role of ET(A)R antagonists in oncologic therapies.


Antineoplastic Agents/pharmacology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Breast Neoplasms/drug therapy , Endothelin-1/metabolism , Metalloendopeptidases/antagonists & inhibitors , Receptors, Endothelin/metabolism , Urogenital Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Aspartic Acid Endopeptidases/metabolism , Breast Neoplasms/metabolism , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Endothelin Receptor Antagonists , Endothelin-Converting Enzymes , Female , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Male , Metalloendopeptidases/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Receptors, Endothelin/agonists , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Urogenital Neoplasms/metabolism , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism
12.
Curr Clin Pharmacol ; 3(2): 85-98, 2008 May.
Article En | MEDLINE | ID: mdl-18690883

In the past few years many encouraging advancements have been made in understanding the molecular mechanisms underlying carcinogenesis and tumor progression. These improvements have led to the identification of promising new targets for cancer therapy. There has been much success with the HER2 targeting antibody trastuzumab (Herceptin) in the treatment of early stage and metastatic breast cancer. Consequently, several antibodies inhibiting cellular signaling of VEGF and EGFR were tested with respect to their efficacy in breast cancer. In phase II and III clinical trials the humanized anti-VEGF antibody bevacizumab (Avastin) alone or in combination with capecitabine exhibited responses in patients with metastatic breast cancer. Recent developments focus on small molecules interfering with different signal transduction pathways in tumor cells. Numerous inhibitors of EGF and VEGF receptor tyrosine kinases and farnesyl transferases are in early stages of clinical development for breast cancer. Another promising approach is the targeting of endothelins and their two G-protein coupled receptors (ET(A)R und ET(B)R). In this article, we will shortly outline well established targeted treatments and discuss the current development of novel agents to be utilized for molecular targeted breast cancer therapy. Due to the heterogeneity of disease and varying response to conventional systemic therapies, these new perceptions may lead to substantial patient benefit and provide a promising basis for future clinical application.


Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Clinical Trials as Topic , Drug Delivery Systems , Female , Genes, erbB-2 , Humans , Neoplasm Proteins/drug effects , Neoplasm Proteins/genetics , Receptor, Endothelin A/drug effects
13.
Cell Oncol ; 29(6): 443-51, 2007.
Article En | MEDLINE | ID: mdl-18032821

BACKGROUND: Recently, we were able to show that amplifications of the epidermal growth factor receptor (egfr) gene and the overexpression of EGFR were associated with the initiation and progression of phyllodes tumours. METHODS: In order to gain further insights into regulation mechanisms associated with egfr amplifications and EGFR expression in phyllodes tumours, we performed global gene expression analysis (Affymetrix A133.2) on a series of 10 phyllodes tumours, of these three with and seven without amplifications of an important regulatory repeat in intron 1 of egfr (CA-SSR I). The results were verified and extended by means of immunohistochemistry using the tissue microarray method on an extensively characterized series of 58 phyllodes tumours with antibodies against caveolin-1, eps15, EGF, TGF-alpha, pErk, pAkt and mdm2. RESULTS: We were able to show that the presence of egfr CA-SSR I amplifications in phyllodes tumours was associated with 230 differentially expressed genes. Caveolin-1 and eps15, involved in EGFR turnover and signalling, were regulated differentially on the RNA and protein level proportionally to egfr gene dosage. Further immunohistochemical analysis revealed that the expression of caveolin-1 and eps15 were also significantly correlated with the expression of pAkt (p<0.05), pERK (p<0.05), mdm2 (p<0.01) and EGF (p<0.001 for caveolin-1). Eps15 and pERK were further associated with tumour grade (p<0.01 and p<0.001, respectively). CONCLUSION: Our results show that amplifications within regulatory sequences of egfr are associated with the expression of eps15 and caveolin-1, indicating an increased turnover of EGFR. The interplay between EGFR and caveolin-1, eps15, pAkt, mdm2 and pERK therefore seems to present a major molecular pathway in carcinogenesis and progression of breast phyllodes tumours.


Breast Neoplasms/genetics , ErbB Receptors/genetics , Gene Amplification , Gene Expression Regulation, Neoplastic , Phyllodes Tumor/genetics , Breast Neoplasms/pathology , Female , Humans , Immunohistochemistry , Mitogen-Activated Protein Kinase 3/genetics , Oligonucleotide Array Sequence Analysis , Phyllodes Tumor/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Neoplasm/genetics , Tissue Array Analysis
14.
World J Surg Oncol ; 5: 110, 2007 Oct 03.
Article En | MEDLINE | ID: mdl-17915016

BACKGROUND: Caveolin-1 is thought to have an important impact on both signal transduction and mediation of intracellular processes. Furthermore, it has been suggested that Caveolin-1 may contribute to certain steps of carcinogenesis in various types of cancer. We examined the potential clinical relevance of Caveolin-1 in normal, benign and malignant breast tissue specimens. METHODS: Using tissue microarray (TMA) technology cases of invasive breast cancer, DCIS, benign breast disease (i.e. fibroadenoma, sclerosing adenosis, ductal hyperplasia and radial scar) and normal breast tissue were evaluated for Caveolin-1 expression. Immunohistochemical staining with an anti-Caveolin-1-antibody was performed. Staining intensity was quantified semiquantitatively. In invasive lesions staining results were correlated with clinical and pathological data. RESULTS: No Caveolin-1 expression was observed in epithelial cells of normal breast tissue (n = 5), benign breast disease (n = 295) and DCIS (n = 108). However, Caveolin-1 expression was found in 32 of 109 cases of invasive breast carcinomas (29.4%). Caveolin-1 expression in invasive breast cancer could neither be correlated with survival parameters such as overall or disease-free survival nor with established clinical and pathological markers. CONCLUSION: In this study we demonstrated expression of Caveolin-1 in one third of invasive breast cancers. A significant increase in Caveolin-1 expression was observed comparing invasive breast cancer to both benign breast tissue and non-invasive breast cancer. Since inhibitors of Caveolin-1 signalling are available, targeting Caveolin-1 in breast cancer may represent a potential option for future breast cancer treatment.

15.
Curr Vasc Pharmacol ; 5(3): 239-48, 2007 Jul.
Article En | MEDLINE | ID: mdl-17627567

The endothelin axis (ET axis), comprising the three peptides endothelin (ET)-1, -2, -3 and their receptors ET(A)R and ET(B)R, is expressed in various cells and tissues. The biologically active ET-1 is formed by endothelin-converting enzyme (ECE) from inactive big-ET-1. ET-1 has emerged as an important peptide in a host of biological functions, including development, cellular proliferation, apoptosis and angiogenesis, thereby playing an important physiological and pathophysiological role. As these effects are mediated by ET(A)R, activation of ET(B)R prevents apoptosis, inhibits ECE expression and mediates the clearance of ET-1. Emerging data indicate that the ET axis is involved in tumourigenesis and tumour progression of various cancers. Expression of the ET axis has been demonstrated in a wide range of human tumours. Since most data have been reported for female malignancies, this review will focus on the role of the ET axis in cancers of the ovary, the cervix and the breast. In ovarian cancer, activation of ET(A)R by ET-1 is a key mechanism in the cellular signalling network promoting cancer growth and progression. Similar effects have been shown for cervical and endometrial cancer. In breast cancer, ET-1 via ET(A)R promotes proliferation and invasion, mediates bone metastases and predicts unfavourable response to chemotherapy. The outstanding role of ET-1 and ET(A)R in carcinogenesis and tumour progression has led to an extensive search for interfering agents, resulting in the development of selective ET(A)R antagonists on the one hand and inhibitors of the endothelin-converting enzyme (ECE) on the other. Targeting the ET axis via ET(A)R or ECE blockade seems to be a promising approach in the treatment of female malignancies.


Antineoplastic Agents/pharmacology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Endothelin A Receptor Antagonists , Endothelin-1/metabolism , Enzyme Inhibitors/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Neoplasms/drug therapy , Amino Acid Sequence , Animals , Antineoplastic Agents/therapeutic use , Aspartic Acid Endopeptidases/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Endothelin B Receptor Antagonists , Endothelin-2/metabolism , Endothelin-3/metabolism , Endothelin-Converting Enzymes , Enzyme Inhibitors/therapeutic use , Female , Humans , Metalloendopeptidases/metabolism , Molecular Sequence Data , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Receptor, Endothelin A/metabolism , Receptor, Endothelin B/metabolism , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism
16.
Oncol Rep ; 18(2): 305-11, 2007 Aug.
Article En | MEDLINE | ID: mdl-17611649

Overexpression of endothelin (ET)-1 and its receptors, ETAR and ETBR, commonly referred to as the 'ET-axis', has been demonstrated to play a role in cancer progression for various human tumours. Based on these results we propose a similar role of the expression of the ET-axis in vulvar cancer. Expression of the ET-axis was investigated immunohistochemically using tissue microarrays with tumour samples of 68 vulvar cancer patients. Samples were obtained from patients undergoing local excision or radical vulvectomy. ET-1 expression of tumour cells correlated highly significantly with early stages of vulvar cancer (p=0.004), whereas neither ETAR nor ETBR expression showed any association with TNM stages. High staining levels of ETBR in the tumour tissue were significantly related to tumour progression (p=0.01) and early metastases (p=0.09); low ETBR staining intensity correlated with longer relapse-free survival (p=0.019). In patients with ETBR overexpressing low-stage tumours (pT1-2) we observed a significantly reduced overall survival and disease-free survival (p=0.036 and 0.021, respectively). ETAR expression and ETBR expression were significantly correlative (p=0.018). Accordingly, co-expression of both receptors was related to tumour progression (p=0.022) and an increased risk for local recurrence (p=0.005). These results suggest that, in addition to established histological and clinical prognostic factors, analysis of ET-receptor and, in particular, of ETBR expression by means of simple immunohistochemical analysis might improve prediction of the prognosis for patients with vulvar squamous cell carcinoma.


Receptor, Endothelin B/biosynthesis , Vulvar Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Disease Progression , Endothelin-1/analysis , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Prognosis , Receptor, Endothelin A/analysis , Survival Analysis , Vulvar Neoplasms/metabolism
17.
Breast Cancer Res Treat ; 106(3): 361-9, 2007 Dec.
Article En | MEDLINE | ID: mdl-17295044

Endothelin-1 (ET-1) and its receptors, ET(A)R and ET(B)R, are overexpressed in breast carcinomas. However, little is known about the relevance of endothelin-converting enzyme-1 (ECE-1) and ET-1 degrading neprilysin (NEP). In this study, expression of ECE-1 and NEP was determined in 600 breast cancer tissue samples by immunohistochemistry; staining results were correlated with clinicopathological parameters. For ECE-1 expression, we found a significant correlation with VEGF (P < 0.001) and ET(A)R expression (P = 0.048). While patients with ECE-1 overexpressing tumours had more frequent disease recurrence (P = 0.03), NEP overexpression correlated with improved disease-free survival (DFS) (P = 0.023) and less frequent metastasis (P = 0.046). Also, a decrease of NEP expression with malignant progression (G1-G3) was found. ECE-1 inhibition using the selective ECE-1 inhibitor RO 67-7447 in MCF-7 breast cancer cells led to a significantly decreased ET-1 expression and reduced cell invasiveness (54.3% of controls, P = 0.014). Our results indicate that overexpression of ECE-1 is associated with unfavourable outcome, whereas NEP positively influences survival. Thus, expression of ECE-1 and NEP may have prognostic relevance. Due to the anti-invasive effect of the selective ECE-1 inhibitor, targeting ECE-1 may represent an innovative option in future breast cancer therapy.


Aspartic Acid Endopeptidases/physiology , Breast Neoplasms/pathology , Metalloendopeptidases/physiology , Neprilysin/physiology , Aspartic Acid Endopeptidases/analysis , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement , Endothelin-1/physiology , Endothelin-Converting Enzymes , Female , Humans , Immunohistochemistry , Metalloendopeptidases/analysis , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Neoplasm Invasiveness , Neprilysin/analysis , Neprilysin/genetics , Prognosis , RNA, Messenger/analysis
18.
Breast Cancer Res ; 9(1): R8, 2007.
Article En | MEDLINE | ID: mdl-17244359

INTRODUCTION: Heparan sulphate proteoglycan syndecan-1 modulates cell proliferation, adhesion, migration and angiogenesis. It is a coreceptor for the hepatocyte growth factor receptor c-met, and its coexpression with E-cadherin is synchronously regulated during epithelial-mesenchymal transition. In breast cancer, changes in the expression of syndecan-1, E-cadherin and c-met correlate with poor prognosis. In this study we evaluated whether coexpression of these functionally linked prognostic markers constitutes an expression signature in ductal carcinoma in situ (DCIS) of the breast that may promote cell proliferation and (lymph)angiogenesis. METHODS: Expression of syndecan-1, E-cadherin and c-met was detected immunohistochemically using a tissue microarray in tumour specimens from 200 DCIS patients. Results were correlated with the expression patterns of angiogenic and lymphangiogenic markers. Coexpression of the three prognostic markers was evaluated in human breast cancer cells by confocal immunofluorescence microscopy and RT-PCR. RESULTS: Coexpression and membrane colocalization of the three markers was confirmed in MCF-7 cells. E-cadherin expression decreased, and c-met expression increased progressively in more aggressive cell lines. Tissue microarray analysis revealed strong positive staining of tumour cells for syndecan-1 in 72%, E-cadherin in 67.8% and c-met in 48.6% of DCIS. E-cadherin expression was significantly associated with c-met and syndecan-1. Expression of c-met and syndecan-1 was significantly more frequent in the subgroup of patients with pure DCIS than in those with DCIS and a coexisting invasive carcinoma. Levels of c-met and syndecan-1 expression were associated with HER2 expression. Expression of c-met significantly correlated with expression of endothelin A and B receptors, vascular endothelial growth factor (VEGF)-A and fibroblast growth factor receptor-1, whereas E-cadherin expression correlated significantly with endothelin A receptor, VEGF-A and VEGF-C staining. CONCLUSION: Syndecan-1, E-cadherin and c-met constitute a marker signature associated with angiogenic and lymphangiogenic factors in DCIS. This coexpression may reflect a state of parallel activation of different signal transduction pathways, promoting tumour cell proliferation and angiogenesis. Our findings have implications for future therapeutic approaches in terms of a multiple target approach, which may be useful early in breast cancer progression.


Cadherins/metabolism , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Lymphangiogenesis , Neovascularization, Pathologic , Syndecan-1/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Intraductal, Noninfiltrating/blood supply , Cell Proliferation , Female , Humans , Proto-Oncogene Proteins c-met/metabolism , Tumor Cells, Cultured
19.
Pathobiology ; 73(1): 18-25, 2006.
Article En | MEDLINE | ID: mdl-16785764

OBJECTIVE: Single nucleotide polymorphism analysis (SNP) has recently been proposed as an alternative technique to comparative genomic hybridization (CGH) for defining loss of heterozygosity and gene copy number changes in a single experimental setup. In order to assess the potential of SNP analysis to complement or, ultimately, substitute CGH results, we applied both techniques to five primary tumor samples and two tumor cell lines. This was complemented by dilution experiments based on normal lymphocyte DNA to decipher the lower detection limit for genetic alterations. METHODS/RESULTS: Using an in-house software tool, we demonstrated that SNP analysis permits the generation of chromosomal alteration patterns that largely resemble conventional CGH ratio profiles of a given tumor/cell line. Moreover, compared to CGH, our SNP software tool allows a much more detailed definition of amplicon sizes and involved candidate genes. This advantage even persisted when the technique was applied to DNA with more than 60% of nontumor content. However, the detection of chromosomal losses is severely hampered by the presence of nontumor DNA so that the use of the SNP technique should be limited to tumor samples with more than 80% tumor DNA. CONCLUSION: SNP analysis is a very valuable tool for the detection and characterization of high-level chromosomal amplifications in the vast majority of primary tumor samples. Our software tool improves the analysis of SNP data and the presentation of the results, bridging the gap to existing CGH knowledge (http://bioinformatics.uni-muenster.de, 'Publications and Supplements').


DNA, Neoplasm/analysis , Gene Dosage , Genetic Techniques , Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Algorithms , Cell Line, Tumor , DNA Mutational Analysis , DNA, Neoplasm/genetics , Female , Gene Amplification , Humans , In Situ Hybridization, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Software
20.
Anticancer Res ; 26(1B): 621-7, 2006.
Article En | MEDLINE | ID: mdl-16739330

BACKGROUND: The overexpression of syndecan-1 in breast carcinomas correlates with poorer prognosis and an aggressive phenotype. The effect of syndecan-1 expression on tumor response to neoadjuvant chemotherapy was determined in locally advanced breast cancer. PATIENTS AND METHODS: Semi-quantitative syndecan-1 immunohistochemistry was performed in pre-chemotherapy breast cancer biopsies of 37 patients undergoing high-dose neoadjuvant treatment with cyclophosphamide and epirubicin. RESULTS: 43.2% of breast carcinomas stained positive for syndecan-1. Syndecan-1 expression was more frequent in ductal invasive carcinomas than in other histological types (p=0.062). The pathological response to chemotherapy was decreased in syndecan-1-positive patients: 37.5% of syndecan-1-positive vs. 19% of syndecan-1-negative patients attained pathologically "no change". No syndecan-1-positive patient showed complete remission. Also, a correlation between syndecan-1 immunostaining intensity and response to chemotherapy was observed. Of the responding tumors, none showed strong syndecan-1 expression (Score 3+), whereas 20% of the non-responding tumors were strongly syndecan-1-positive. CONCLUSION: Syndecan-1-expressing breast carcinomas show a trend towards a decreased response to chemotherapy.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/metabolism , Membrane Glycoproteins/biosynthesis , Proteoglycans/biosynthesis , Adult , Aged , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Chemotherapy, Adjuvant , Cyclophosphamide/administration & dosage , Epirubicin/administration & dosage , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Humans , Immunohistochemistry , Middle Aged , Neoadjuvant Therapy , Predictive Value of Tests , Syndecan-1 , Syndecans
...