Network


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

Hotspot


Dive into the research topics where Pia Herrmann is active.

Publication


Featured researches published by Pia Herrmann.


PLOS ONE | 2012

Circulating MACC1 Transcripts in Colorectal Cancer Patient Plasma Predict Metastasis and Prognosis

Ulrike Stein; Susen Burock; Pia Herrmann; Ina Wendler; Markus Niederstrasser; Klaus D. Wernecke; Peter M. Schlag

Background Metastasis is the most frequent cause of treatment failure and death in colorectal cancer. Early detection of tumors and metastases is crucial for improving treatment strategies and patient outcome. Development of reliable biomarkers and simple tests routinely applicable in the clinic for detection, prognostication, and therapy monitoring is of special interest. We recently identified the novel gene Metastasis-Associated in Colon Cancer 1 (MACC1), a key regulator of the HGF/Met-pathway. MACC1 is a strong prognostic biomarker for colon cancer metastasis and allows identification of high-risk subjects in early stages, when determined in patients’ primary tumors. To overcome the limitation of a restricted number of molecular analyses in tumor tissue, the establishment of a non-invasive blood test for early identification of high-risk cancer patients, for monitoring disease course and therapy response is strongly needed. Methodology/Principal Findings For the first time, we describe a non-invasive assay for quantification of circulating MACC1 transcripts in blood of more than 300 colorectal cancer patients. MACC1 transcript levels are increased in all disease stages of the cancer patients compared to tumor-free volunteers. Highest MACC1 levels were determined in individuals with metastases (all P<0.05). Importantly, high MACC1 levels correlate with unfavorable survival (P<.0001). Combining MACC1 with circulating transcripts of the metastasis gene S100A4, a transcriptional target of the Wnt/β-catenin-pathway, improves survival prediction for newly diagnosed cancer patients. Conclusion/Significance This blood-based assay for circulating MACC1 transcripts, which can be quantitated on a routine basis, is clinically applicable for diagnosis, prognosis, and therapeutic monitoring of cancer patients. Here we demonstrate the diagnostic and prognostic value of circulating MACC1 transcripts in patient plasma for metastasis and survival. Since MACC1 represents a promising target for anti-metastatic therapies, circulating MACC1 transcripts may prove to be an ideal read-out for monitoring therapeutic response of future interventions targeting MACC1-induced metastasis in cancer patients.


Annals of Surgery | 2012

Integrative marker analysis allows risk assessment for metastasis in stage II colon cancer.

Ulrich Nitsche; Robert D. Rosenberg; Alexander Balmert; Tibor Schuster; Julia Slotta-Huspenina; Pia Herrmann; Franz G. Bader; Helmut Friess; Peter M. Schlag; Ulrike Stein; Klaus-Peter Janssen

Objectives:Individualized risk assessment in patients with UICC stage II colon cancer based on a panel of molecular genetic alterations. Background:Risk assessment in patients with colon cancer and localized disease (UICC stage II) is not sufficiently reliable. Development of metachronous metastasis is assumed to be governed largely by individual tumor genetics. Methods:Fresh frozen tissue from 232 patients (T3-4, N0, M0) with complete tumor resection and a median follow-up of 97 months was analyzed for microsatellite stability, KRAS exon 2, and BRAF exon 15 mutations. Gene expression of the WNT-pathway surrogate marker osteopontin and the metastasis-associated genes SASH1 and MACC1 was determined for 179 patients. The results were correlated with metachronous distant metastasis risk (n = 22 patients). Results:Mutations of KRAS were detected in 30% patients, mutations of BRAF in 15% patients, and microsatellite instability in 26% patients. Risk of recurrence was associated with KRAS mutation (P = 0.033), microsatellite stable tumors (P = 0.015), decreased expression of SASH1 (P = 0.049), and increased expression of MACC1 (P < 0.001). MACC1 was the only independent parameter for recurrence prediction (hazard ratio: 6.2; 95% confidence interval: 2.4–16; P < 0.001). Integrative 2-step cluster analysis allocated patients into 4 groups, according to their tumor genetics. KRAS mutation, BRAF wild type, microsatellite stability, and high MACC1 expression defined the group with the highest risk of recurrence (16%, 7 of 43), whereas BRAF wild type, microsatellite instability, and low MACC1 expression defined the group with the lowest risk (4%, 1 of 26). Conclusions:MACC1 expression predicts development of metastases, outperforming microsatellite stability status, as well as KRAS/BRAF mutation status.


BMC Cancer | 2015

Heterogeneity analysis of Metastasis Associated in Colon Cancer 1 (MACC1) for survival prognosis of colorectal cancer patients: a retrospective cohort study.

Viktor H. Koelzer; Pia Herrmann; Inti Zlobec; Eva Karamitopoulou; Alessandro Lugli; Ulrike Stein

BackgroundMetastasis of colorectal cancer (CRC) is directly linked to patient survival. We previously identified the novel gene Metastasis Associated in Colon Cancer 1 (MACC1) in CRC and demonstrated its importance as metastasis inducer and prognostic biomarker. Here, we investigate the geographic expression pattern of MACC1 in colorectal adenocarcinoma and tumor buds in correlation with clinicopathological and molecular features for improvement of survival prognosis.MethodsWe performed geographic MACC1 expression analysis in tumor center, invasive front and tumor buds on whole tissue sections of 187 well-characterized CRCs by immunohistochemistry. MACC1 expression in each geographic zone was analyzed with Mismatch repair (MMR)-status, BRAF/KRAS-mutations and CpG-island methylation.ResultsMACC1 was significantly overexpressed in tumor tissue as compared to normal mucosa (p < 0.001). Within colorectal adenocarcinomas, a significant increase of MACC1 from tumor center to front (p = 0.0012) was detected. MACC1 was highly overexpressed in 55% tumor budding cells. Independent of geographic location, MACC1 predicted advanced pT and pN-stages, high grade tumor budding, venous and lymphatic invasion (p < 0.05). High MACC1 expression at the invasive front was decisive for prediction of metastasis (p = 0.0223) and poor survival (p = 0.0217). The geographic pattern of MACC1 did not correlate with MMR-status, BRAF/KRAS-mutations or CpG-island methylation.ConclusionMACC1 is differentially expressed in CRC. At the invasive front, MACC1 expression predicts best aggressive clinicopathological features, tumor budding, metastasis formation and poor survival outcome.


World Journal of Gastroenterology | 2015

Circulating metastasis associated in colon cancer 1 transcripts in gastric cancer patient plasma as diagnostic and prognostic biomarker

Susen Burock; Pia Herrmann; Ina Wendler; Markus Niederstrasser; Klaus-Dieter Wernecke; Ulrike Stein

AIM To evaluate the diagnostic and prognostic value of circulating Metastasis Associated in Colon Cancer 1 (MACC1) transcripts in plasma of gastric cancer patients. METHODS We provide for the first time a blood-based assay for transcript quantification of the metastasis inducer MACC1 in a prospective study of gastric cancer patient plasma. MACC1 is a strong prognostic biomarker for tumor progression and metastasis in a variety of solid cancers. We conducted a study to define the diagnostic and prognostic power of MACC1 transcripts using 76 plasma samples from gastric cancer patients, either newly diagnosed with gastric cancer, newly diagnosed with metachronous metastasis of gastric cancer, as well as follow-up patients. Findings were controlled by using plasma samples from 54 tumor-free volunteers. Plasma was separated, RNA was isolated, and levels of MACC1 as well as S100A4 transcripts were determined by quantitative RT-PCR. RESULTS Based on the levels of circulating MACC1 transcripts in plasma we significantly discriminated tumor-free volunteers and gastric cancer patients (P < 0.001). Levels of circulating MACC1 transcripts were increased in gastric cancer patients of each disease stage, compared to tumor-free volunteers: patients with tumors without metastasis (P = 0.005), with synchronous metastasis (P = 0.002), with metachronous metastasis (P = 0.005), and patients during follow-up (P = 0.021). Sensitivity was 0.68 (95%CI: 0.45-0.85) and specificity was 0.89 (95%CI: 0.77-0.95), respectively. Importantly, gastric cancer patients with high circulating MACC1 transcript levels in plasma demonstrated significantly shorter survival when compared with patients demonstrating low MACC1 levels (P = 0.0015). Furthermore, gastric cancer patients with high circulating transcript levels of MACC1 as well as of S100A4 in plasma demonstrated significantly shorter survival when compared with patients demonstrating low levels of both biomarkers or with only one biomarker elevated (P = 0.001). CONCLUSION Levels of circulating MACC1 transcripts in plasma of gastric cancer patients are of diagnostic value and are prognostic for patient survival in a prospective study.


Molecular Cancer | 2015

High MACC1 expression in combination with mutated KRAS G13 indicates poor survival of colorectal cancer patients.

Katharina Ilm; Wolfgang Kemmner; Marc Osterland; Susen Burock; Gudrun Koch; Pia Herrmann; Peter M. Schlag; Ulrike Stein

BackgroundThe metastasis-associated in colon cancer 1 (MACC1) gene has been identified as prognostic biomarker for colorectal cancer (CRC). Here, we aimed at the refinement of risk assessment by separate and combined survival analyses of MACC1 expression with any of the markers KRAS mutated in codon 12 (KRAS G12) or codon 13 (KRAS G13), BRAF V600 mutation and MSI status in a retrospective study of 99 CRC patients with tumors UICC staged I, II and III.FindingsWe showed that only high MACC1 expression (HR: 6.09, 95% CI: 2.50-14.85, P < 0.001) and KRAS G13 mutation (HR: 5.19, 95% CI: 1.06-25.45, P = 0.042) were independent prognostic markers for shorter metastasis-free survival (MFS). Accordingly, Cox regression analysis revealed that patients with high MACC1 expression and KRAS G13 mutation exhibited the worst prognosis (HR: 14.48, 95% CI: 3.37-62.18, P < 0.001). Patients were classified based on their molecular characteristics into four clusters with significant differences in MFS (P = 0.003) by using the SPSS 2-step cluster function and Kaplan-Meier survival analysis.ConclusionAccording to our results, patients with high MACC1 expression and mutated KRAS G13 exhibited the highest risk for metachronous metastases formation. Moreover, we demonstrated that the “Traditional pathway” with an intermediate risk for metastasis formation can be further subdivided by assessing MACC1 expression into a low and high risk group with regard to MFS prognosis. This is the first report showing that identification of CRC patients at high risk for metastasis is possible by assessing MACC1 expression in combination with KRAS G13 mutation.


Hepatology | 2015

Metastasis-associated in colon cancer 1 is an independent prognostic biomarker for survival in klatskin tumor patients

Andri Lederer; Pia Herrmann; Daniel Seehofer; Manfred Dietel; Johann Pratschke; Peter M. Schlag; Ulrike Stein

Curative treatment of intrahepatic cholangiocarcinoma (ICC) and hilar cholangiocarcinoma (Klatskin tumors) is limited to surgical resection or orthotopic liver transplantation. However, not all patients benefit from a surgical approach and suffer from early tumor recurrence. Response to chemotherapy is generally poor and, until today, no targeted therapy could be established. Metastasis‐associated in colon cancer 1 (MACC1) is a recently discovered regulator of the hepatocyte growth factor (HGF)/Met/mitogen‐activated protein kinase pathway, which induces proliferation, migration, and invasion in cell culture, as well as metastasis in mice. MACC1 expression shows a significant correlation with Met expression in colon cancer tissue and is highly prognostic for occurrence of distant metastasis and survival in colon cancer patients. Thus, we aimed to measure the expression of MACC1, Met, and HGF messenger RNA in microdissected tumor tissue and corresponding normal liver tissue of 156 patients with Klatskin tumors (n = 76) and ICC (n = 80) using real‐time quantitative reverse‐transcriptase polymerase chain reaction. We used immunohistochemical staining to validate the results. MACC1 expression in tumor tissue of both tumor entities was significantly higher than in corresponding normal liver tissue (P < 0.001). Klatskin tumor patients with a history of tumor recurrence had significantly higher MACC1 expression than those without tumor recurrence (P = 0.005). Uni‐ und multivariate survival analysis showed that Klatskin tumor patients with high MACC1 had a significantly shorter overall (OS) and disease‐free survival (DFS; P = 0.001 and P < 0.001, respectively). The multivariate analysis confirmed MACC1 to be an independent factor for overall survival in Klatskin tumor patients (hazard ratio: 2.777; 95% confidence interval: 1.389‐5.555; P = 0.004). Conclusion: Our study identified MACC1 as a highly prognostic biomarker for OS and DFS in Klatskin tumor patients. MACC1 expression could become an important diagnostic tool and might be a candidate for targeted therapy. (Hepatology 2015;62:841–850)


Cancer Research | 2017

Abstract 2778: Combination of DNA mismatch repair status and MACC1 expression in patients with stage II colon cancer: The BIOGRID studies

Ulrich-Peter Rohr; Pia Herrmann; Katharina Ilm; Hai Zhang; Sabine Lohmann; Astrid Reiser; Andrea Muranyi; Janice Smith; Susen Burock; Marc Osterland; Katherine Leith; Shalini Singh; Patrick Brunhoeber; Rebecca Bowermaster; Jeanne Tie; Michael Christie; Hui-Li Wong; Paul Waring; Kandavel Shanmugam; Peter Gibbs; Ulrike Stein

We have previously identified the gene Metastasis-Associated in Colon Cancer 1 (MACC1). MACC1 acts a prognostic biomarker for tumor progression, metastasis and patient survival for a broad variety of solid cancer types. Here we assessed if the MACC1 gene could separate stage II colon cancer patients with proficient mismatch repair (pMMR) into high- and low-risk groups who might benefit from or be spared adjuvant chemotherapy based on their prognosis. In the Charite 1 discovery cohort (n=61), MACC1 expression and MSI status were assayed by qRT-PCR in cryo-preserved tumors from CRC patients. MSS/MSI-low/MACC1-low tumors showed better survival vs. MSS/MSI-low/MACC1-high (Pl0.0001). Patients with MSS/MSI-low/MACC1-low tumors had a similar prognosis as patients with MSI-H tumors. The Charite 2 comparison cohort (n=40) was used to translate MACC1 qRT-PCR analyses to FFPE samples. MACC1 expression was significantly higher in metachronously metastasizing tumors linked to shorter relapse-free survival (RFS), independent of the tissue type analyzed (cryo-preserved or FFPE). Next we translated MACC1 mRNA levels from qRT-PCR to MACC1 protein levels from immunohistochemistry (IHC) by comparing them in consecutive FFPE tumor sections in the BIOGRID 1 training cohort (n=189) enriched for disease recurrence. Chemotherapy-naive patients with unfavorable pMMR status separated into MACC1-high and -low groups. Better RFS was seen in the pMMR/MACC1-low vs. pMMR/MACC1-high group using MACC1 mRNA and protein expression. pMMR/MACC1-low expression was seen in 12% and 8% of patients by qRT-PCR and IHC; interestingly, they had the same favorable prognosis as the deficient MMR (dMMR) group. Prognostic and predictive findings from BIOGRID 1 were confirmed in the independent BIOGRID 2 validation cohort (n=306) unenriched for recurrence. Better RFS was again seen in chemotherapy-naive patients in the pMMR/MACC1-low (6%) vs. pMMR/MACC1-high group. No patients with pMMR/MACC1-low phenotype had disease recurrence. Remarkably, pooling BIOGRID 1 and 2, 5-year RFS was 100% and thus significantly longer in the pMMR/MACC1-low vs. pMMR/MACC1-high group (P=0.037). Taken together, MACC1 expression, measured by qRT-PCR or IHC, differentiates patients with unfavorable pMMR status. Patients with stage II colon cancer and pMMR/MACC1-low tumor status have a similar favorable prognosis as those patients with dMMR status who might not benefit from adjuvant therapy. Citation Format: Ulrich-Peter Rohr, Pia Herrmann, Katharina Ilm, Hai Zhang, Sabine Lohmann, Astrid Reiser, Andrea Muranyi, Janice Smith, Susen Burock, Marc Osterland, Katherine Leith, Shalini Singh, Patrick Brunhoeber, Rebecca Bowermaster, Jeanne Tie, Michael Christie, Hui-Li Wong, Paul Waring, Kandavel Shanmugam, Peter Gibbs, Ulrike S. Stein. Combination of DNA mismatch repair status and MACC1 expression in patients with stage II colon cancer: The BIOGRID studies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2778. doi:10.1158/1538-7445.AM2017-2778


Cancer Research | 2014

Abstract 45: MACC1 expression and KRas13 mutation for improved survival prognosis of colorectal cancer patients

Katharina Ilm; Wolfgang Kemmner; Gudrun Koch; Pia Herrmann; Marc Osterland; Senji Shirasawa; Takehiko Sasazuki; Ulrike Stein

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Colorectal cancer (CRC) is the third most common cancer worldwide. The metastatic dissemination of the primary tumor is directly linked to patient survival. We previously discovered the gene metastasis-associated in colon cancer 1 (MACC1). MACC1 has been identified as a new remarkable biomarker for disease prognosis and prediction of therapy response for CRC and for a variety of solid cancers. MACC1 expression in tumor tissues and patient blood correlates with tumor formation and progression, development of metastases and patient survival representing a decisive driver for tumorigenesis and metastasis. Here we aim at the refinement of risk assessment in a retrospective study of 100 CRC patients with tumors staged I, II and III by combinatorial analyses of MACC1 with the KRas and BRaf mutation status. First, we correlated the single parameters, KRas12, KRas13, BRaf600 mutation and MACC1 expression with clinical outcome data, such as metastasis-free survival (MFS). We found correlations of high MACC1 expression with shorter MFS (P<0.001). Likewise, patients with a somatic KRas13 mutation showed decreased MFS (P=0.003). Interestingly, KRas12 or BRaf600 mutations as well as their wild type counterparts had no impact on survival prognosis. Multivariate analyses combining MACC1 expression level with the single parameters KRas12, KRas13 and BRaf600 mutation revealed an improved prognosis exclusively for KRas13 mutated tumors vs. MACC1 expression alone. High MACC1 expressers with KRas13 mutation had the poorest median MFS (17.2 months) compared to MACC1 high/KRas wild type patients (37.2 months; P=0.039). Further, we addressed the impact of KRas13 on regulation of MACC1 expression in vitro. We used RNAi to analyze the impact of KRas signaling on MACC1 in several CRC cell lines with different KRas mutation status. In addition, we employed HCT116 cells harboring the activating G13D KRAS mutation together with the HCT116-derived sublines Hkh-2 and Hke-3 lacking the mutated KRas13 allele by homologous recombination. Disrupting the constitutive active KRas signaling led to increased MACC1 mRNA and protein expression levels in HCT116 cells, either by knock-down of the endogenous G13D mutated KRas expression by RNAi or through gene targeting. In summary, MACC1 expression levels together with KRas13 mutation status was the only combination that improved the identification of CRC patients at high risk for metastasis formation and the prediction of MFS when compared to MACC1 alone Citation Format: Katharina Ilm, Wolfgang Kemmner, Gudrun Koch, Pia Herrmann, Marc Osterland, Senji Shirasawa, Takehiko Sasazuki, Ulrike S. Stein. MACC1 expression and KRas13 mutation for improved survival prognosis of colorectal cancer patients. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 45. doi:10.1158/1538-7445.AM2014-45


Blood | 2005

Nongenotoxic activation of the p53 pathway as a therapeutic strategy for multiple myeloma

Thorsten Stühmer; Manik Chatterjee; Martin Hildebrandt; Pia Herrmann; Hella Gollasch; Christian Gerecke; Sebastian Theurich; Luisa Cigliano; Rudolf A. Manz; Peter T. Daniel; Kurt Bommert; Lyubomir T. Vassilev; Ralf C. Bargou


Blood | 2002

In the presence of bone marrow stromal cells human multiple myeloma cells become independent of the IL-6/gp130/STAT3 pathway

Manik Chatterjee; Dirk Honemann; Suzanne Lentzsch; Kurt Bommert; Christine Sers; Pia Herrmann; Stephan Mathas; Bernd Dörken; Ralf C. Bargou

Collaboration


Dive into the Pia Herrmann's collaboration.

Top Co-Authors

Avatar

Ulrike Stein

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Janice Smith

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Marc Osterland

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Mathias Dahlmann

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ina Wendler

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Katharina Ilm

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Mario Löhr

University of Würzburg

View shared research outputs
Researchain Logo
Decentralizing Knowledge