Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Saskia von Stillfried is active.

Publication


Featured researches published by Saskia von Stillfried.


Epigenetics | 2014

Epigenetic inactivation of the novel candidate tumor suppressor gene ITIH5 in colon cancer predicts unfavorable overall survival in the CpG island methylator phenotype

Vera Kloten; Michael Rose; Sophie Kaspar; Saskia von Stillfried; Ruth Knüchel; Edgar Dahl

Inter-α-trypsin inhibitor heavy chain 5 (ITIH5) is supposed to be involved in extracellular matrix stability and thus may play a key role in the inhibition of tumor progression. The current study is the first to analyze in depth ITIH5 expression as well as its potential clinical and functional impact in colon cancer. Based on 30 tumor and 30 adjacent normal tissues we examined ITIH5 mRNA expression and promoter methylation, whose significance was further validated by independent data sets from The Cancer Genome Atlas (TCGA) platform. In addition, ITIH5 protein expression was evaluated using immunohistochemistry. ITIH5 mRNA expression loss was significantly associated (P < 0.001) with hypermethylation of the ITIH5 promoter in primary colon tumors. In addition, treatment of tumor cell lines with demethylating (DAC) and histone acetylating (TSA) agents induced ITIH5 expression. In line, independent TCGA data revealed a significant expression loss of ITIH5, particularly in the MSI-high and CIMP-positive phenotype concordant with an increased ITIH5 hypermethylation in CIMP-positive colon tumors (P < 0.001). In proximal, i.e., right-sided tumors, abundant ITIH5 expression was associated with longer overall survival (OS, P = 0.049) and the CIMP-positive (P = 0.032) subgroup. Functionally, ITIH5 re-expression mediated a reduced proliferation in HCT116 and CaCo2 cells. In conclusion, our results indicate that ITIH5 is a novel putative tumor suppressor gene in colon cancer with a potential impact in the CIMP-related pathway. ITIH5 may serve as a novel epigenetic-based diagnostic biomarker with further clinical impact for risk stratification of CIMP-positive colon cancer patients.


PLOS ONE | 2015

AGR3 in breast cancer: prognostic impact and suitable serum-based biomarker for early cancer detection.

Stefan Garczyk; Saskia von Stillfried; Wiebke Antonopoulos; Arndt Hartmann; Michael G. Schrauder; Peter A. Fasching; Tobias Anzeneder; Andrea Tannapfel; Yavuz H. Ergönenc; Ruth Knüchel; Michael Rose; Edgar Dahl

Blood-based early detection of breast cancer has recently gained novel momentum, as liquid biopsy diagnostics is a fast emerging field. In this study, we aimed to identify secreted proteins which are up-regulated both in tumour tissue and serum samples of breast cancer patients compared to normal tissue and sera. Based on two independent tissue cohorts (n = 75 and n = 229) and one serum cohort (n = 80) of human breast cancer and healthy serum samples, we characterised AGR3 as a novel potential biomarker both for breast cancer prognosis and early breast cancer detection from blood. AGR3 expression in breast tumours is significantly associated with oestrogen receptor α (P<0.001) and lower tumour grade (P<0.01). Interestingly, AGR3 protein expression correlates with unfavourable outcome in low (G1) and intermediate (G2) grade breast tumours (multivariate hazard ratio: 2.186, 95% CI: 1.008-4.740, P<0.05) indicating an independent prognostic impact. In sera analysed by ELISA technique, AGR3 protein concentration was significantly (P<0.001) elevated in samples from breast cancer patients (n = 40, mainly low stage tumours) compared to healthy controls (n = 40). To develop a suitable biomarker panel for early breast cancer detection, we measured AGR2 protein in human serum samples in parallel. The combined AGR3/AGR2 biomarker panel achieved a sensitivity of 64.5% and a specificity of 89.5% as shown by receiver operating characteristic (ROC) curve statistics. Thus our data clearly show the potential usability of AGR3 and AGR2 as biomarkers for blood-based early detection of human breast cancer.


Angiogenesis | 2016

Contrast-enhanced CT imaging in patients with chronic kidney disease

Saskia von Stillfried; Jonas C. Apitzsch; Josef Ehling; Tobias Penzkofer; Andreas H. Mahnken; Ruth Knüchel; Jürgen Floege; Peter Boor

Renal microvascular rarefaction characterizes chronic kidney disease (CKD). In murine models of CKD, micro-CT imaging reflected capillary rarefaction using quantification of renal relative blood volume (rBV). In addition, micro-CT imaging revealed morphological alterations of the intrarenal vasculature including reduced vascular branching and lumen diameter. Here, we retrospectively quantified rBV in contrast-enhanced CT angiography in patients and found that, compared to non-CKD patients, those with CKD and renal fibrosis had significantly reduced rBV in the renal cortex. rBV values closely mirrored capillary rarefaction in the corresponding nephrectomy specimens. In patients with follow-up CT angiography, reduction of renal function was paralleled by a decline in rBV. Using virtual autopsy, i.e., postmortem CT angiography, morphometry of intrarenal arteries in 3D-rendered CT images revealed significantly reduced arterial diameter and branching in CKD compared to non-CKD cases. In conclusion, in CKD patients, contrast-enhanced CT imaging with quantification of rBV correlates with functional renal vasculature, whereas virtual autopsy allows morphometric analyses of macrovascular changes. Importantly, the observed vascular alterations in CKD patients mirror those in animals with progressive CKD, suggesting a high relevance of animal models for studying vascular alterations in CKD and renal fibrosis.


PLOS ONE | 2016

Loss of Dickkopf 3 Promotes the Tumorigenesis of Basal Breast Cancer

Eva Lorsy; Aylin Sophie Topuz; Cordelia Geisler; Sarah Stahl; Stefan Garczyk; Saskia von Stillfried; Mareike Hoss; Oleg Gluz; Arndt Hartmann; Ruth Knüchel; Edgar Dahl

Dickkopf 3 (DKK3) has been associated with tumor suppression of various tumor entities including breast cancer. However, the functional impact of DKK3 on the tumorigenesis of distinct molecular breast cancer subtypes has not been considered so far. Therefore, we initiated a study analyzing the subtype-specific DKK3 expression pattern as well as its prognostic and functional impact with respect to breast cancer subtypes. Based on three independent tissue cohorts including one in silico dataset (n = 30, n = 463 and n = 791) we observed a clear down-regulation of DKK3 expression in breast cancer samples compared to healthy breast tissue controls on mRNA and protein level. Interestingly, most abundant reduction of DKK3 expression was detected in the highly aggressive basal breast cancer subtype. Analyzing a large in silico dataset comprising 3,554 cases showed that low DKK3 mRNA expression was significantly associated with reduced recurrence free survival (RFS) of luminal and basal-like breast cancer cases. Functionally, DKK3 re-expression in human breast cancer cell lines led to suppression of cell growth possibly mediated by up-regulation of apoptosis in basal-like but not in luminal-like breast cancer cell lines. Moreover, ectopic DKK3 expression in mesenchymal basal breast cancer cells resulted in partial restoration of epithelial cell morphology which was molecularly supported by higher expression of epithelial markers like E-Cadherin and down-regulation of mesenchymal markers such as Snail 1. Hence, we provide evidence that down-regulation of DKK3 especially promotes tumorigenesis of the aggressive basal breast cancer subtype. Further studies decoding the underlying molecular mechanisms of DKK3-mediated effects may help to identify novel targeted therapies for this clinically highly relevant breast cancer subtype.


bioRxiv | 2017

Detection of small breast tumors using tumor penetrating-polymersomes engineered to target p32 protein

Pablo Scodeller; Desirè Di Silvio; Sergio Salzar Fuentes; Vanessa Gómez Vallejo; Xabier Ríos; Eneko San Sebastián; Meina Suck; Federica De Lorenzi; Larissa Y. Rizzo; Saskia von Stillfried; Kalle Kilk; Twan Lammers; Sergio Moya; Tambet Teesalu

Triple negative breast cancer (TNBC) is the deadliest form of breast cancer and its successful treatment critically depends on early diagnosis and therapy. The multi-compartment protein p32 is overexpressed and present at cell surfaces in a variety of tumors, including TNBC, specifically in the malignant cells and endothelial cells, and in macrophages localized in hypoxic areas of the tumor. Herein we used polyethylene glycol-polycaprolactone polymersomes that were affinity targeted with the p32-binding tumor penetrating peptide LinTT1 (AKRGARSTA) for imaging of TNBC lesions. A tyrosine residue was added to the peptide to allow for 124I labeling and PET imaging. In a TNBC model in mice, systemic LinTT1-targeted polymersomes accumulated in early tumor lesions more than twice as efficiently as untargeted polymersomes with up to 20% ID/cc at 24 h after administration. The PET-imaging was very sensitive, allowing detection of tumors as small as ~20mm3. Confocal imaging of tumor tissue sections revealed a high degree of vascular exit and stromal penetration of LinTT1-targeted polymersomes and co-localization with tumor-associated macrophages. Our studies show that systemic LinTT1-targeted polymersomes can be potentially used for precision-guided tumor imaging and treatment of TNBC.Triple negative breast cancer (TNBC) is the deadliest form of breast cancer and its successful treatment critically depends on early diagnosis and therapy. The multi-compartment protein p32 is overexpressed and present at cell surfaces in TNBC, specifically in the malignant cells and endothelial cells, and in macrophages localized in hypoxic areas of the tumor. Herein we used polyethylene glycol-polycaprolactone polymersomes that were affinity targeted with the p32-binding tumor penetrating peptide LinTT1 (AKRGARSTA) for imaging of TNBC lesions. A tyrosine residue was added to the peptide to allow for 124I labeling and PET imaging. Systemic LinTT1-targeted polymersomes accumulated in early tumor lesions more than twice as efficiently as untargeted polymersomes with up to 20% ID/cc at 24 h after administration. The PET-imaging was very sensitive, allowing detection of tumors as small as ~20mm3. Confocal imaging of tumor tissue sections revealed a high degree of vascular exit and stromal penetration of LinTT1-targeted polymersomes and co-localization with tumor-associated macrophages. Our studies show that systemic LinTT1-targeted polymersomes can be potentially used for precision-guided tumor imaging and treatment of TNBC. uFigure1 Small triple negative brast tumors can be detected with LinTT1-conjugated polymersomes. Radiolabeled LinTT1-polymersomes were intravenously injected into mice bearing small triple negative breast tumor. LinTT1 peptide binds to p32 protein expressed in the surface of tumor cells and activated macrophage/myeloid cells. LinTT1 is cleaved by urokinase type plasminogen activator (uPA) in tumor, and the processed peptide binds to NRP-1, triggering the penetration of polymersomes into the tumor tissue.


Oncotarget | 2017

Oncogenic features of neuromedin U in breast cancer are associated with NMUR2 expression involving crosstalk with members of the WNT signaling pathway

Stefan Garczyk; Natalie Klotz; Sabrina Szczepanski; Bernd Denecke; Wiebke Antonopoulos; Saskia von Stillfried; Ruth Knüchel; Michael R. Rose; Edgar Dahl

Neuromedin U (NMU) has been shown driving the progression of various tumor entities, including breast cancer. However, the expression pattern of NMU and its receptors in breast cancer tissues as well as systematic insight into mechanisms and downstream targets of the NMU-driven signaling pathways are still elusive. Here, NMU expression was found up-regulated in all breast cancer subtypes when compared to healthy breast tissue. Using an in silico dataset comprising 1,195 samples, high NMU expression was identified as an indicator of poor outcome in breast tumors showing strong NMUR2 expression. Next, the biological impact of NMU on breast cancer cells in relation to NMUR2 expression was analyzed. Ectopic NMU expression reduced colony growth while promoting a motile phenotype in NMUR2-positive SKBR3 but not NMUR2-negative Hs578T cells. To uncover signaling pathways and key molecules affected by NMU in SKBR3 cells, Affymetrix microarray analysis was applied. Forced NMU expression affected molecules involved in WNT receptor signaling among others. As such we demonstrated enhanced activation of the WNT/planar cell polarity (PCP) effector RAC1 and down-regulation of canonical WNT targets such as MYC. In summary, NMU might contribute to progression of NMUR2-positive breast cancer representing a potential druggable target for future personalized strategies.


Molecular Imaging and Biology | 2016

Multimodal imaging of breast cancer metastasis targeting and antimetastatic nanotherapy

Larissa Y. Rizzo; Cristianne J.F. Rijcken; Robert Pola; Gert Storm; Josef Ehling; Saskia von Stillfried; Fabian Kiessling; Twan Lammers

BODY: Abstract Body: Introduction: Spectroscopic photoacoustic (sPA) imaging allows for identification of individual optical absorbers within biological tissues, endogenously limited to hemoglobin, melanin, and lipids. Molecularly specific contrast agents are needed to investigate other tissue markers, but few agents are considered readily clinically translatable. Our objective was to explore the potentially clinically translatable combination of antibodies (Ab) and the FDA-approved near-infrared, fluorescent dye indocyanine green (ICG) for sPA imaging of CD276, a molecular marker differentially expressed in breast cancer1, which may improve accuracy of ultrasound imaging for breast cancer detection. Methods: Succinimidyl ester modified ICG was conjugated to monoclonal Ab specific to the CD276 marker. Ab-dye binding ratios were determined using spectrophotometric analysis and protein concentration was determined with a standard BCA assay. A transgenic mouse model for breast cancer development (FVB/N Tg(MMTV/PyMT634Mul) was used to assess the ability of sPA imaging to detect the accumulation of CD276-ICG contrast agent in breast cancer. Mice with invasive breast adenocarcinoma (10-12 weeks of age) were injected intravenously with 33 μg of CD276-ICG or control agents, including isotype Ab conjugated with ICG (Iso-ICG), ICG dye alone, or CD276-ICG in tumor negative mice. Fluorescence, multi-wavelength (680-950 nm, 10 nm increments) sPA, and B-mode ultrasound imaging were performed before and 24h, 48h, and 72h after i.v. administration using the VisualSonics LAZR and the Xenogen IVIS Spectrum. Anatomical B-mode images were used to guide ROI selection for sPA data analysis. Using an in-house sPA data analysis algorithm, the average molecular CD276 signal in the tumor ROI was determined by monitoring absorbance shifts of ICG. CD276-ICG uptake and clearance were monitored with fluorescence imaging. Immunohistochemical (IHC) staining was used to quantify CD276 expression in breast cancer and normal mammary gland tissue. Results: In total, 80 tumors were imaged over the five day period with another 110 monitored with various control agents (Iso-ICG (n= 30), CD276-ICG in normal mammary glands (n = 60), and ICG only (n=20). Tumors showed a 3.15±0.42 fold increase in molecular CD276 signal compared to pre-injection values with sPA (range 0.30–20.0 fold increase, p<0.001) and a 1.37±0.15 fold increase in CD276 signal with fluorescence imaging (range 0.75–2.02 fold increase, p<0.001). Control agent values showed no significant increase in signal compared to background values. Murine breast cancer tissue, both epithelial and endothelial cells, stained positive for CD276, while normal tissue did not express CD276. Conclusions: Spectroscopic photoacoustic imaging is able to detect clinically translatable antibody-dye contrast agents in a transgenic mouse model of breast cancer. CD276-targeted molecular sPA signals were detected as early as 24 hours and for at least 96 hours after injection. Molecular sPA imaging may become a complementary parameter to ultrasound imaging for more accurate earlier detection of breast cancer. 1. Seaman S., et. al. Cancer Cell. 2007;11(6):539-554 AUTHORS (LAST NAME, FIRST NAME): Wilson, Katheryne1; Bachawal, Sunitha1; Willmann, Juergen K.1 INSTITUTIONS (ALL): 1. Radiology, Stanford University, Stanford, CA, United States. (No Image Selected) DOI: 10.1007/s11307-01 -0 Mol Imaging Biol (201 ) 1 (Suppl 1): 1 1553 S 6 8 Y


Oncotarget | 2018

Application of polymersomes engineered to target p32 protein for detection of small breast tumors in mice

Pablo Scodeller; Sergio Salazar Fuentes; Vanessa Gómez Vallejo; Xabier Ríos; Eneko San Sebastián; Valeria Sidorenko; Desirè Di Silvio; Meina Suck; Federica De Lorenzi; Larissa Y. Rizzo; Saskia von Stillfried; Kalle Kilk; Twan Lammers; Sergio Moya; Tambet Teesalu

Triple negative breast cancer (TNBC) is the deadliest form of breast cancer and its successful treatment critically depends on early diagnosis and therapy. The multi-compartment protein p32 is overexpressed and present at cell surfaces in a variety of tumors, including TNBC, specifically in the malignant cells and endothelial cells, and in macrophages localized in hypoxic areas of the tumor. Herein we used polyethylene glycol-polycaprolactone polymersomes that were affinity targeted with the p32-binding tumor penetrating peptide LinTT1 (AKRGARSTA) for imaging of TNBC lesions. A tyrosine residue was added to the peptide to allow for 124I labeling and PET imaging. In a TNBC model in mice, systemic LinTT1-targeted polymersomes accumulated in early tumor lesions more than twice as efficiently as untargeted polymersomes with up to 20% ID/cc at 24 h after administration. The PET-imaging was very sensitive, allowing detection of tumors as small as ∼20 mm3. Confocal imaging of tumor tissue sections revealed a high degree of vascular exit and stromal penetration of LinTT1-targeted polymersomes and co-localization with tumor-associated macrophages. Our studies show that systemic LinTT1-targeted polymersomes can be potentially used for precision-guided tumor imaging and treatment of TNBC.


Angiogenesis | 2016

In situ validation of VEGFR-2 and α v ß 3 integrin as targets for breast lesion characterization

Josef Ehling; Matthias Misiewicz; Saskia von Stillfried; Diana Möckel; Jessica Bzyl; Sibylle Pochon; Wiltrud Lederle; Ruth Knuechel; Twan Lammers; Moritz Palmowski; Fabian Kiessling


CardioVascular and Interventional Radiology | 2017

Development and Evaluation of a Novel Curved Biopsy Device for CT-Guided Biopsy of Lesions Unreachable Using Standard Straight Needle Trajectories

M Schulze-Hagen; Jochen Pfeffer; Markus Zimmermann; M Liebl; Saskia von Stillfried; Christiane K. Kuhl; Philipp Bruners; Peter Isfort

Collaboration


Dive into the Saskia von Stillfried's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edgar Dahl

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge