Network


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

Hotspot


Dive into the research topics where Bernd Neumaier is active.

Publication


Featured researches published by Bernd Neumaier.


Journal of Clinical Oncology | 1999

Early Detection and Accurate Description of Extent of Metastatic Bone Disease in Breast Cancer With Fluoride Ion and Positron Emission Tomography

Holger Schirrmeister; Albrecht Guhlmann; Jörg Kotzerke; Claudia Santjohanser; Thorsten Kühn; Rolf Kreienberg; Peter M. Messer; Karin Nüssle; Klaus Elsner; Gerhard Glatting; Harald Träger; Bernd Neumaier; Christoph G. Diederichs; Sven N. Reske

PURPOSE Previous studies have shown that bone metastases are revealed by magnetic resonance imaging (MRI) or bone marrow scintigraphy several months before they are visible by conventional bone scintigraphy (BS). We present a new approach for detecting bone metastases in patients with breast cancer. We compared findings obtained with fluoride ion (F-18) and positron emission tomography (PET) with those obtained with conventional BS. PATIENTS AND METHODS Thirty-four breast cancer patients were prospectively examined using F-18-PET and conventional BS. F-18-PET and BS were performed within 3 weeks of each other. Metastatic bone disease was previously known to be present in six patients and was suspected (bone pain or increasing levels of tumor markers, Ca(2+), alkaline phosphatase) in 28 patients. Both imaging modalities were compared by patient-by-patient analysis and lesion-by-lesion analysis, using a five-point scale for receiver operating characteristic (ROC) curve analysis. A panel of reference methods was used, including MRI (28 patients), planar x-ray (17 patients), and spiral computed tomography (four patients). RESULTS With F-18-PET, 64 bone metastases were detected in 17 patients. Only 29 metastases were detected in 11 patients with BS. As a result of F-18-PET imaging, clinical management was changed in four patients (11.7%). For F-18-PET, the area under the ROC curve was 0.99 on a lesion basis (for BS, it was 0.74; P <.05) and 1.00 on a patient basis (for BS, it was 0.82; P <.05). CONCLUSION F-18-PET demonstrates a very early bone reaction when small bone marrow metastases are present, allowing accurate detection of breast cancer bone metastases. This accurate detection has a significant effect on clinical management, compared with the effect on management brought about by detection with conventional BS.


Molecular Imaging and Biology | 2015

Comparison of [18F]DCFPyL and [68Ga]Ga-PSMA-HBED-CC for PSMA-PET Imaging in Patients with Relapsed Prostate Cancer

Markus Dietlein; Carsten Kobe; Georg Kuhnert; Simone Stockter; Thomas Fischer; Klaus Schomäcker; Matthias Schmidt; Felix Dietlein; Boris D. Zlatopolskiy; Philipp Krapf; Raphael Richarz; Stephan Neubauer; Alexander Drzezga; Bernd Neumaier

PurposeGallium-68 (Ga-68)-labeled tracers for imaging expression of the prostate-specific membrane antigen (PSMA) such as the [68Ga]Ga-PSMA-HBED-CC have already demonstrated high potential for the detection of recurrent prostate cancer. However, compared to Ga-68, a labeling with fluorine-18 (F-18) would offer advantages with respect to availability, production amount, and image resolution. [18F]DCFPyL is a promising F-18-labeled candidate for PSMA-positron emission tomography (PET) imaging that has been recently introduced. In the current study, we aimed to compare [68Ga]Ga-PSMA-HBED-CC and [18F]DCFPyL for clinical use in biochemically relapsed prostate cancer.ProceduresIn 14 selected patients with PSA relapse of prostate cancer, [18F]DCFPyL PET/X-ray computed tomography (CT) was performed in addition to [68Ga]Ga-PSMA-HBED-CC PET/CT. A systematic comparison was carried out between results obtained with both tracers with regard to the number of detected PSMA-positive lesions, the standardized uptake value (SUV)max and the lesion to background ratios.ResultsAll suspicious lesions identified by [68Ga]Ga-PSMA-HBED-CC were also detected with [18F]DCFPyL. In three patients, additional lesions were observed using [18F]DCFPyL PET/CT. The mean SUVmax in the concordant [18F]DCFPyL PSMA-positive lesions was significantly higher as compared to [68Ga]Ga-PSMA-HBED-CC (14.5 vs. 12.2, p = 0.028, n = 15). The mean tumor to background ratios (n = 15) were significantly higher for [18F]DCFPyL compared to [68Ga]Ga-PSMA-HBED-CC using kidney, spleen, or parotid as reference organs (p = 0.006, p = 0.002, p = 0.008), but no significant differences were found using the liver (p = 0.167) or the mediastinum (p = 0.363) as reference organs.Conclusion[18F]DCFPyL PET/CT provided a high image quality and visualized small prostate lesions with excellent sensitivity. [18F]DCFPyL represents a highly promising alternative to [68Ga]Ga-PSMA-HBED-CC for PSMA-PET/CT imaging in relapsed prostate cancer.


Neuroreport | 2000

In vivo imaging of activated microglia using [11C]PK11195 and positron emission tomography in patients after ischemic stroke.

Alexander Gerhard; Bernd Neumaier; Ercan Elitok; Gerhard Glatting; Vincent Ries; Reinhard Tomczak; Albert C. Ludolph; Sven N. Reske

Neuroprotective strategies are currently being developed for stroke patients. Although the focus is on the development of early treatment the importance of late pathogenetic events is increasingly recognized. To investigate the microglial reaction in stroke we used a marker for activated microglia, [11C]PK11195, and PET in five patients with ischemic stroke 5–53 days after infarction. In one patient serial measurements were made. We demonstrated in each individual and at each point in time that a microglial reaction takes place in the area where T1 weighted MRI (magnetic resonance imaging) shows intensity changes. We consider this PET method as a promising tool to study the late pathogenetic consequences of cerebral infarction and to evaluate neuroprotective strategies with respect to the consequences of the microglial activation.


Clinical Cancer Research | 2008

Glioma Proliferation as Assessed by 3‘-Fluoro-3’-Deoxy-l-Thymidine Positron Emission Tomography in Patients with Newly Diagnosed High-Grade Glioma

Roland T. Ullrich; Heiko Backes; Hongfeng Li; Lutz W. Kracht; Hrvoje Miletic; Kristina Kesper; Bernd Neumaier; Wolf-Dieter Heiss; Klaus Wienhard; Andreas H. Jacobs

Purpose: The aim of this study was to investigate the relationship between the in vivo derived kinetic parameters of 3′-deoxy-3′-18F-fluorothymidine (18F-FLT) and the proliferation rate measured in vitro by Ki-67 staining in patients with newly diagnosed high-grade gliomas. Experimental Design: Thirteen patients with newly diagnosed high-grade gliomas were investigated with 18F-FLT and methyl-11C- l-methionine (11C-MET) positron emission tomography (PET) and T1-, Gd-T1–, and T2-weighted magnetic resonance imaging on consecutive days. Tracer kinetic parameters of 18F-FLT as well as the standardized uptake value and the tumor-to-background (T/B) ratio of 18F-FLT and 11C-MET were determined. Data of kinetic modeling, standardized uptake value, and T/B values derived from 18F-FLT-PET were compared with T/B values derived from 11C-MET-PET and to the in vitro proliferation marker Ki-67. Results: A significant correlation was observed between the metabolic rate constant Ki and the proliferation index as measured by Ki-67 immunostaining [Ki, r = 0.79 (P = 0.004)]. Also, the phosphorylation rate constant k3 correlated with Ki-67 [k3, r = 0.76 (P = 0.006)], whereas the rate constant for transport through the blood brain barrier K1 showed a weaker correlation with Ki-67 [K1, r = 0.62 (P = 0.044)]. No significant correlation between 11C-MET and 18F-FLT uptake ratios and Ki-67 was observed. Conclusions: This study shows that kinetic analysis of 18F-FLT tracer uptake is essential for the in vivo assessment of tumor proliferation in high-grade gliomas, whereas uptake ratios of 11C-MET and 18F-FLT failed to correlate with the in vitro determined proliferation marker. Thus, kinetic analysis of 18F-FLT might provide an accurate method for the assessment of early response to glioma treatment in the future.


Journal of Clinical Investigation | 2013

Tumor VEGF:VEGFR2 autocrine feed-forward loop triggers angiogenesis in lung cancer

Sampurna Chatterjee; Lukas C. Heukamp; Maike Siobal; Jakob Schöttle; Caroline Wieczorek; Martin Peifer; Davide Frasca; Mirjam Koker; Katharina König; Lydia Meder; Daniel Rauh; Reinhard Buettner; Johanna Wolf; Rolf A. Brekken; Bernd Neumaier; Gerhard Christofori; Roman K. Thomas; Roland T. Ullrich

The molecular mechanisms that control the balance between antiangiogenic and proangiogenic factors and initiate the angiogenic switch in tumors remain poorly defined. By combining chemical genetics with multimodal imaging, we have identified an autocrine feed-forward loop in tumor cells in which tumor-derived VEGF stimulates VEGF production via VEGFR2-dependent activation of mTOR, substantially amplifying the initial proangiogenic signal. Disruption of this feed-forward loop by chemical perturbation or knockdown of VEGFR2 in tumor cells dramatically inhibited production of VEGF in vitro and in vivo. This disruption was sufficient to prevent tumor growth in vivo. In patients with lung cancer, we found that this VEGF:VEGFR2 feed-forward loop was active, as the level of VEGF/VEGFR2 binding in tumor cells was highly correlated to tumor angiogenesis. We further demonstrated that inhibition of tumor cell VEGFR2 induces feedback activation of the IRS/MAPK signaling cascade. Most strikingly, combined pharmacological inhibition of VEGFR2 (ZD6474) and MEK (PD0325901) in tumor cells resulted in dramatic tumor shrinkage, whereas monotherapy only modestly slowed tumor growth. Thus, a tumor cell-autonomous VEGF:VEGFR2 feed-forward loop provides signal amplification required for the establishment of fully angiogenic tumors in lung cancer. Interrupting this feed-forward loop switches tumor cells from an angiogenic to a proliferative phenotype that sensitizes tumor cells to MAPK inhibition.


Journal of Cerebral Blood Flow and Metabolism | 2009

Neuroinflammation Extends Brain Tissue at Risk to Vital Peri-Infarct Tissue: A Double Tracer [11C]PK11195- and [18F]FDG-PET Study

Michael Schroeter; Maria Adele Dennin; Maureen Walberer; Heiko Backes; Bernd Neumaier; Gereon R. Fink; Rudolf Graf

Focal cerebral ischemia elicits strong inflammatory responses involving activation of resident microglia and recruitment of monocytes/macrophages. These cells express peripheral benzodiazepine receptors (PBRs) and can be visualized by positron emission tomography (PET) using [11C]PK11195 that selectively binds to PBRs. Earlier research suggests that transient ischemia in rats induces increased [11C]PK11195 binding within the infarct core. In this study, we investigated the expression of PBRs during permanent ischemia in rats. Permanent cerebral ischemia was induced by injection of macrospheres into the middle cerebral artery. Multimodal imaging 7 days after ischemia comprised (1) magnetic resonance imaging that assessed the extent of infarcts; (2) [18F]-2-fluoro-2-deoxy-d-glucose ([18F]FDG)-PET characterizing cerebral glucose transport and metabolism; and (3) [11C]PK11195-PET detecting neuroinflammation. Immunohistochemistry verified ischemic damage and neuroinflammatory processes. Contrasting with earlier data for transient ischemia, no [11C]PK11195 binding was found in the infarct core. Rather, permanent ischemia caused increased [11C]PK11195 binding in the normoperfused peri-infarct zone (mean standard uptake value (SUV): 1.93 ± 0.49), colocalizing with a 60% increase in the [18F]FDG metabolic rate constant with accumulated activated microglia and macrophages. These results suggest that after permanent focal ischemia, neuroinflammation occurring in the normoperfused peri-infarct zone goes along with increased energy demand, therefore extending the tissue at risk to areas adjacent to the infarct.


British Journal of Haematology | 2005

188Re or 90Y-labelled anti-CD66 antibody as part of a dose-reduced conditioning regimen for patients with acute leukaemia or myelodysplastic syndrome over the age of 55: results of a phase I–II study

Mark Ringhoffer; Norbert M. Blumstein; Bernd Neumaier; Gerhard Glatting; Stephanie von Harsdorf; Inga Buchmann; Markus Wiesneth; Jörg Kotzerke; Thorsten Zenz; Andreas K. Buck; Peter Schauwecker; Stephan Stilgenbauer; Hartmut Döhner; Sven N. Reske; Donald Bunjes

In a phase I–II study for patients aged 55–65 years, we employed radioimmunotherapy using an anti‐CD‐66 antibody as part of a dose‐reduced conditioning regimen, which was followed by a T‐cell‐depleted graft. 20 patients with a median age of 63 years suffering from acute leukaemia (n = 17) or myelodysplastic syndrome (n = 3) received the antibody labelled either with 188Rhenium (n = 8) or with 90Yttrium (n = 12) during conditioning. Radioimmunotherapy provided a mean dose of 21·9 (±8·4) Gy to the bone marrow with a significantly higher dose when 90Yttrium was used. Additional conditioning was fludarabine‐based plus anti‐thymocyte globulin in matched related donor transplants (n = 11), or plus melphalan in matched unrelated donor transplants (n = 9). Regimen‐related toxicity was low, with two patients developing three episodes of grade III organ toxicity. All patients engrafted, grade II–IV acute graft‐versus‐host disease (GvHD) was observed in one patient (5%) and chronic GvHD in three patients (15%). The cumulative incidence of non‐relapse mortality was 25%, the cumulative incidence of relapse 55%. The probability of survival was estimated to be 70% at 1 year and 52% at 2 years post‐transplant, although no plateau was reached afterwards. In conclusion, radioimmunotherapy using the anti‐CD66 antibody was feasible and safe in our elderly patient group and provided a high marrow dose.


The Journal of Neuroscience | 2010

Noninvasive Imaging of Endogenous Neural Stem Cell Mobilization In Vivo Using Positron Emission Tomography

Maria Adele Rueger; Heiko Backes; Maureen Walberer; Bernd Neumaier; Roland T. Ullrich; Marie-Lune Simard; Beata Emig; Gereon R. Fink; Mathias Hoehn; Rudolf Graf; Michael Schroeter

Neural stem cells reside in two major niches in the adult brain [i.e., the subventricular zone (SVZ) and the dentate gyrus of the hippocampus]. Insults to the brain such as cerebral ischemia result in a physiological mobilization of endogenous neural stem cells. Since recent studies showed that pharmacological stimulation can be used to expand the endogenous neural stem cell niche, hope has been raised to enhance the brains own regenerative capacity. For the evaluation of such novel therapeutic approaches, longitudinal and intraindividual monitoring of the endogenous neural stem cell niche would be required. However, to date no conclusive imaging technique has been established. We used positron emission tomography (PET) and the radiotracer 3′-deoxy-3′-[18F]fluoro-l-thymidine ([18F]FLT) that enables imaging and measuring of proliferation to noninvasively detect endogenous neural stem cells in the normal and diseased adult rat brain in vivo. This method indeed visualized neural stem cell niches in the living rat brain, identified as increased [18F]FLT-binding in the SVZ and the hippocampus. Focal cerebral ischemia and subsequent damage of the blood–brain barrier did not interfere with the capability of [18F]FLT-PET to visualize neural stem cell mobilization. Moreover, [18F]FLT-PET allowed for an in vivo quantification of increased neural stem cell mobilization caused by pharmacological stimulation or by focal cerebral ischemia. The data suggest that noninvasive longitudinal monitoring and quantification of endogenous neural stem cell activation in the brain is feasible and that [18F]FLT-PET could be used to monitor the effects of drugs aimed at expanding the neural stem cell niche.


PLOS ONE | 2008

Early Detection of Erlotinib Treatment Response in NSCLC by 3′-Deoxy-3′-[18F]-Fluoro-L-Thymidine ([18F]FLT) Positron Emission Tomography (PET)

Roland T. Ullrich; Thomas Zander; Bernd Neumaier; Mirjam Koker; Takeshi Shimamura; Yannic Waerzeggers; Christa L. Borgman; Samir Tawadros; Hongfeng Li; Martin L. Sos; Heiko Backes; Geoffrey I. Shapiro; Jürgen Wolf; Andreas H. Jacobs; Roman K. Thomas; Alexandra Winkeler

Background Inhibition of the epidermal growth factor receptor (EGFR) has shown clinical success in patients with advanced non-small cell lung cancer (NSCLC). Somatic mutations of EGFR were found in lung adenocarcinoma that lead to exquisite dependency on EGFR signaling; thus patients with EGFR-mutant tumors are at high chance of response to EGFR inhibitors. However, imaging approaches affording early identification of tumor response in EGFR-dependent carcinomas have so far been lacking. Methodology/Principal Findings We performed a systematic comparison of 3′-Deoxy-3′-[18F]-fluoro-L-thymidine ([18F]FLT) and 2-[18F]-fluoro-2-deoxy-D-glucose ([18F]FDG) positron emission tomography (PET) for their potential to identify response to EGFR inhibitors in a model of EGFR-dependent lung cancer early after treatment initiation. While erlotinib-sensitive tumors exhibited a striking and reproducible decrease in [18F]FLT uptake after only two days of treatment, [18F]FDG PET based imaging revealed no consistent reduction in tumor glucose uptake. In sensitive tumors, a decrease in [18F]FLT PET but not [18F]FDG PET uptake correlated with cell cycle arrest and induction of apoptosis. The reduction in [18F]FLT PET signal at day 2 translated into dramatic tumor shrinkage four days later. Furthermore, the specificity of our results is confirmed by the complete lack of [18F]FLT PET response of tumors expressing the T790M erlotinib resistance mutation of EGFR. Conclusions [18F]FLT PET enables robust identification of erlotinib response in EGFR-dependent tumors at a very early stage. [18F]FLT PET imaging may represent an appropriate method for early prediction of response to EGFR TKI treatment in patients with NSCLC.


The Journal of Nuclear Medicine | 2011

Quantitative analysis of response to treatment with Erlotinib in advanced non–small cell lung cancer using 18F-FDG and 39-deoxy-39-18F-fluorothymidine PET

Deniz Kahraman; Matthias Scheffler; Thomas Zander; Lucia Nogova; Adriaan A. Lammertsma; Ronald Boellaard; Bernd Neumaier; Roland T. Ullrich; Arne Holstein; Markus Dietlein; Jürgen Wolf; Carsten Kobe

The purpose of this study was to evaluate the relevance for the prediction of clinical benefit of first-line treatment with erlotinib using different quantitative parameters for PET with both 18F-FDG and 3′-deoxy-3′-18F-fluorothymidine (18F-FLT) in patients with advanced non–small cell lung cancer. Methods: Data were used from a prospective trial involving patients with untreated stage IV non–small cell lung cancer. 18F-FDG PET and 18F-FLT PET were performed before and 1 (early) and 6 (late) weeks after erlotinib treatment. Several quantitative standardized uptake values (SUVs) using different definitions of volumes of interest with varying isocontours (maximum SUV [SUVmax], 2-dimensional peak SUV [SUV2Dpeak], 3-dimensional [3D] peak SUV [SUV3Dpeak], 3D isocontour at 50% of the maximum pixel value [SUV50], 3D isocontour at 50% adapted for background [SUVA50], 3D isocontour at 41% of the maximum pixel value adapted for background [SUVA41], 3D isocontour at 70% of the maximum pixel value [SUV70], 3D isocontour at 70% adapted for background [SUVA70], and relative SUV threshold level [SUVRTL]) and metabolically active volume measurements were obtained in the hottest single tumor lesion and in the sum of up to 5 lesions per scan in 30 patients. Metabolic response was defined as a minimum reduction of 30% in each of the different SUVs and as a minimum reduction of 45% in metabolically active volume. Progression-free survival (PFS) was compared between patients with and without metabolic response measured with each of the different parameters, using Kaplan–Meier statistics and a log-rank test. Results: Patients with a metabolic response on early 18F-FDG PET and 18F-FLT PET in the hottest single tumor lesion as well as in the sum of up to 5 lesions per scan had a significantly longer PFS, regardless of the method used to calculate SUV. However, the highest significance was obtained for SUVmax, SUV50, SUVA50, and SUVA41. Patients with a metabolic response measured by SUVmax and SUV3Dpeak on late 18F-FDG PET in the hottest single tumor lesion had a significantly longer PFS. Furthermore, Kaplan–Meier analyses showed a strong association between PFS and response seen by metabolically active volume, measured either in early 18F-FLT or in late 18F-FDG. Conclusion: Early 18F-FDG PET and 18F-FLT PET can predict PFS regardless of the method used for SUV calculation. However, SUVmax, SUV50, SUVA50, and SUVA41 measured with 18F-FDG might be the best robust SUV to use for early response prediction. Metabolically active volume measurement in early 18F-FLT PET and late 18F-FDG PET may have an additional predictive value in monitoring response in patients with advanced non–small cell lung cancer treated with erlotinib.

Collaboration


Dive into the Bernd Neumaier's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Felix M. Mottaghy

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge