156 results on '"Auernhammer CJ"'
Search Results
2. Therapie und Diagnostik von neuroendokrinen Tumoren in der Chirurgischen Klinik der Universität München-Großhadern: Analyse von Inzidenz, Lokalisation und Behandlung über 15 Jahre
- Author
-
Zahn, S, Fertmann, JM, Auernhammer, CJ, Spitzweg, C, Hornung, H, Pöpperl, G, Schick, KS, Jauch, KW, and Hoffmann, JN
- Subjects
ddc: 610 ,610 Medical sciences ,Medicine - Abstract
Einleitung: Bei neuroendokrinen Tumoren (NET) handelt es sich um seltene Tumoren, welche sich bezüglich ihres biologischen Verhaltens erheblich von Tumoren epithelialen Ursprungs unterscheiden. NET können in Behandlungszentren interdisziplinär durch ein breites therapeutisches Armentarium[for full text, please go to the a.m. URL], 126. Kongress der Deutschen Gesellschaft für Chirurgie
- Published
- 2009
- Full Text
- View/download PDF
3. Spezifische c-Met Inhibition hat keine direkten antiproliferativen Effekte auf humane neuroendokrine Tumorzelllinien in vitro
- Author
-
Reuther, C, primary, Heinzle, V, additional, Spampatti, M, additional, Vlotides, G, additional, de Toni, E, additional, Spöttl, G, additional, Maurer, J, additional, Göke, B, additional, and Auernhammer, CJ, additional
- Published
- 2014
- Full Text
- View/download PDF
4. Antiproliferative Effekte des MDM-2 Inhibitors CGM-097 auf humane neuroendokrine GOT1 Tumorzellen in vitro
- Author
-
Reuther, C, primary, Heinzle, V, additional, Nölting, S, additional, Spöttl, G, additional, Maurer, J, additional, Göke, B, additional, and Auernhammer, CJ, additional
- Published
- 2014
- Full Text
- View/download PDF
5. mTOR Inhibition durch Metformin - antiproliferative Effekte auf neuroendokrine Tumorzellen in vitro
- Author
-
Vlotides, G, primary, Tanyeri, A, additional, Spampatti, MP, additional, Zitzmann, K, additional, Spöttl, G, additional, Maurer, J, additional, Nölting, S, additional, Göke, B, additional, and Auernhammer, CJ, additional
- Published
- 2013
- Full Text
- View/download PDF
6. mTOR Inhibition durch Aspirin - antiproliferative Effekte auf neuroendokrine Tumorzellen in vitro
- Author
-
Spampatti, MP, primary, Vlotides, G, additional, Spöttl, G, additional, Maurer, J, additional, Göke, B, additional, and Auernhammer, CJ, additional
- Published
- 2013
- Full Text
- View/download PDF
7. Das neue Somatostatin/Dopamin-Chimär BIM-23A758 induziert in der Midgutcarcinoid-Zelllinie GOT1 potente antitumorale Effekte
- Author
-
Zitzmann, K, primary, Andersen, S, additional, Brand, S, additional, Culler, MD, additional, Lichtl, J, additional, Spöttl, G, additional, Göke, B, additional, and Auernhammer, CJ, additional
- Published
- 2009
- Full Text
- View/download PDF
8. Präsidentenposter: Duale Inhibition der PI(3)K-Akt-mTOR und der Ras-Raf-MEK-Erk1/2 Signalkaskade in neuroendokrinen Tumorzelllinien pankreatischen-, midgut- und bronchialen Ursprungs
- Author
-
Zitzmann, K, primary, Rüden, J von, additional, Brand, S, additional, Göke, B, additional, Lichtl, J, additional, Spöttl, G, additional, and Auernhammer, CJ, additional
- Published
- 2009
- Full Text
- View/download PDF
9. Zielgerichtete molekulare Therapie von neuroendokrinen Tumoren mittels Isoform-spezifischer Akt Inhibition
- Author
-
Zitzmann, K, primary, Brand, S, additional, Göke, B, additional, Meinecke, J, additional, Spöttl, G, additional, and Auernhammer, CJ, additional
- Published
- 2008
- Full Text
- View/download PDF
10. Differences in the antiviral properties of the novel IL-10 like cytokines IL-26, IL-28A/B, and IL-29 against hepatitis C virus infection
- Author
-
Diegelmann, J, primary, Beigel, F, additional, Zitzmann, K, additional, Kaul, A, additional, Göke, B, additional, Auernhammer, CJ, additional, Bartenschlager, R, additional, Diepolder, H, additional, and Brand, S, additional
- Published
- 2008
- Full Text
- View/download PDF
11. Increased expression of the novel IL-10 like cytokine IL-26 in RORγt expressing Th17 cells in patients with active Crohn's disease
- Author
-
Diegelmann, J, primary, Auernhammer, CJ, additional, Göke, B, additional, and Brand, S, additional
- Published
- 2008
- Full Text
- View/download PDF
12. IL-22 mediated liver cell regeneration is abrogated by SOCS-1/3 overexpression
- Author
-
Dambacher, J, primary, Beigel, F, additional, Zitzmann, K, additional, Olszak, T, additional, Prüfer, T, additional, Steib, CJ, additional, Storr, M, additional, Göke, B, additional, Diepolder, H, additional, Bilzer, M, additional, Auernhammer, CJ, additional, and Brand, S, additional
- Published
- 2006
- Full Text
- View/download PDF
13. The novel IL-10 related cytokine IL-26 is increased in active inflammatory bowel disease and intestinal epithelial cells express the functional IL-26 receptor complex
- Author
-
Dambacher, J, primary, Beigel, F, additional, Zitzmann, K, additional, Eichhorst, ST, additional, Otte, J, additional, Seiderer, J, additional, Diepolder, H, additional, Auernhammer, CJ, additional, Ochsenkühn, T, additional, Goeke, B, additional, and Brand, S, additional
- Published
- 2006
- Full Text
- View/download PDF
14. The novel Lambda-interferons IL-28A and IL-29 inhibit HCV replication in vitro and hepatic IL-28 mRNA expression is increased in HCV and CMV infection in vivo
- Author
-
Beigel, F, primary, Dambacher, J, additional, Zitzmann, K, additional, Storr, M, additional, Olszak, T, additional, Goeke, B, additional, Auernhammer, CJ, additional, Kaul, A, additional, Bartenschlager, R, additional, Diepolder, H, additional, and Brand, S, additional
- Published
- 2006
- Full Text
- View/download PDF
15. Characterization of the promoter region of murine NNT-1/BSF-3– a novel paracrine modulator of corticotroph function
- Author
-
Zitzmann, K, primary, Vlotides, G, additional, and Auernhammer, CJ, additional
- Published
- 2005
- Full Text
- View/download PDF
16. PACAP and VIP induce expression of the novel neurotrophin-1/B-cell stimulating factor-3 (Cardiotrophin-like cytokine) in pituitary folliculostellate TtT/GF cells: downstream signaling events involve PKA, PKC and ERK1/2
- Author
-
Vlotides, G, primary, Zitzmann, K, additional, Engelhardt, D, additional, Spoettl, G, additional, Meinecke, J, additional, Stalla, GK, additional, and Auernhammer, CJ, additional
- Published
- 2004
- Full Text
- View/download PDF
17. The oncostatin M receptor/gp130 ligand murine oncostatin M induces apoptosis in adrenocortical Y-1 tumor cells
- Author
-
Auernhammer, CJ, primary, Dorn, F, additional, Vlotides, G, additional, Hengge, S, additional, Kopp, FB, additional, Spoettl, G, additional, Cengic, N, additional, Engelhardt, D, additional, and Weber, MM, additional
- Published
- 2004
- Full Text
- View/download PDF
18. Effects of growth hormone and insulin-like growth factor I binding to natural killer cells
- Author
-
Bidlingmaier, M., primary, Auernhammer, CJ, additional, Feldmeier, H., additional, and Strasburger, CJ, additional
- Published
- 1997
- Full Text
- View/download PDF
19. Compensatory activation of Akt in response to mTOR and Raf inhibitors - a rationale for dual-targeted therapy approaches in neuroendocrine tumor disease.
- Author
-
Zitzmann K, Rüden J, Brand S, Göke B, Lichtl J, Spöttl G, and Auernhammer CJ
- Published
- 2010
- Full Text
- View/download PDF
20. Validation of the standardization framework SSTR-RADS 1.0 for neuroendocrine tumors using the novel SSTR‑targeting peptide [ 18 F]SiTATE.
- Author
-
Ebner R, Lohse A, Fabritius MP, Rübenthaler J, Wängler C, Wängler B, Schirrmacher R, Völter F, Schmid HP, Unterrainer LM, Öcal O, Hinterberger A, Spitzweg C, Auernhammer CJ, Geyer T, Ricke J, Bartenstein P, Holzgreve A, and Grawe F
- Subjects
- Humans, Female, Male, Middle Aged, Reproducibility of Results, Aged, Adult, Aged, 80 and over, Octreotide analogs & derivatives, Fluorine Radioisotopes, Neuroendocrine Tumors diagnostic imaging, Positron Emission Tomography Computed Tomography methods, Positron Emission Tomography Computed Tomography standards, Receptors, Somatostatin metabolism, Radiopharmaceuticals
- Abstract
Objectives: Somatostatin receptor positron emission tomography/computed tomography (SSTR-PET/CT) using [
68 Ga]-labeled tracers is a widely used imaging modality for neuroendocrine tumors (NET). Recently, [18 F]SiTATE, a SiFAlin tagged [Tyr3]-octreotate (TATE) PET tracer, has shown great potential due to favorable clinical characteristics. We aimed to evaluate the reproducibility of Somatostatin Receptor-Reporting and Data System 1.0 (SSTR-RADS 1.0) for structured interpretation and treatment planning of NET using [18 F]SiTATE., Methods: Four readers assessed [18 F]SiTATE-PET/CT of 95 patients according to the SSTR-RADS 1.0 criteria at two different time points. Each reader evaluated up to five target lesions per scan. The overall scan score and the decision on peptide receptor radionuclide therapy (PRRT) were considered. Inter- and intra-reader agreement was determined using the intraclass correlation coefficient (ICC)., Results: The ICC analysis on the inter-reader agreement using SSTR-RADS 1.0 for identical target lesions (ICC ≥ 85%), overall scan score (ICC ≥ 90%), and the decision to recommend PRRT (ICC ≥ 85%) showed excellent agreement. However, significant differences were observed in recommending PRRT among experienced readers (ER) (p = 0.020) and inexperienced readers (IR) (p = 0.004). Compartment-based analysis demonstrated good to excellent inter-reader agreement for most organs (ICC ≥ 74%), except for lymph nodes (ICC ≥ 53%)., Conclusion: SSTR-RADS 1.0 represents a highly reproducible and consistent framework system for stratifying SSTR-targeted PET/CT scans, even using the novel SSTR-ligand [18 F]SiTATE. Some inter-reader variability was observed regarding the evaluation of uptake intensity prior to PRRT as well as compartment scoring of lymph nodes, indicating that those categories require special attention during further clinical validation and might be refined in a future SSTR-RADS version 1.1., Clinical Relevance Statement: SSTR-RADS 1.0 is a consistent framework for categorizing somatostatin receptor-targeted PET/CT scans when using [18 F]SiTATE. The framework serves as a valuable tool for facilitating and improving the management of patients with NET., Key Points: SSTR-RADS 1.0 is a valuable tool for managing patients with NET. SSTR-RADS 1.0 categorizes patients with showing strong agreement across diverse reader expertise. As an alternative to [68 Ga]-labeled PET/CT in neuroendocrine tumor imaging, SSTR-RADS 1.0 reliably classifies [18 F]SiTATE-PET/CT., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
21. PET/CT imaging of differentiated and medullary thyroid carcinoma using the novel SSTR-targeting peptide [ 18 F]SiTATE - first clinical experiences.
- Author
-
Kunte SC, Wenter V, Toms J, Lindner S, Unterrainer M, Eilsberger F, Jurkschat K, Wängler C, Wängler B, Schirrmacher R, Tiling MW, Sheikh GT, Mehrens D, Brendel M, Rübenthaler J, Auernhammer CJ, Spitzweg C, Unterrainer LM, and Holzgreve A
- Abstract
Purpose: The novel
18 F-labeled somatostatin receptor (SSTR)-directed radiotracer [18 F]SiTATE demonstrated promising results for the imaging of various SSTR-expressing tumor types. Although thyroid carcinomas (TC) express SSTR, data on [18 F]SiTATE PET/CT imaging in TC are lacking. This study explores the use of [18 F]SiTATE PET/CT in a patient cohort with histologically proven TC., Methods: As part of a prospective observational study at a single tertiary cancer center, 21 patients with TC (10 medullary (MTC) and 11 differentiated (DTC)) who underwent at least one [18 F]SiTATE PET/CT were included (37 scans in total). Mean SUVmax and SUVmean of tumoral lesions, mean total-tumor-volume (TTV), and whole-body (WB)-SUVmax and WB-SUVmean on PET with their standard deviations (SDs) were determined. PET parameters were correlated to clinical parameters including tumor marker levels (thyroglobulin for DTC, calcitonin for MTC)., Results: 89 lesions were included in the analysis. Metastases were localized in the bone, lymph nodes, lung, soft tissue, and thyroid bed. Osseous (31 lesions; SUVmax 8.6 ± 8.0; SUVmean 5.8 ± 5.4) and nodal (37 lesions; SUVmax 8.7 ± 7.8; SUVmean 5.7 ± 5.4) metastases showed the highest uptake. The MTC disease burden on PET significantly correlated with the calcitonin tumor marker level (e.g., TTV: r = 0.771, r2 = 0.594, p = 0.002). For DTC, no such correlation was present., Conclusion: Our data demonstrate high feasibility of [18 F]SiTATE PET/CT in a small cohort of patients with MTC and DTC. The use of [18 F]SiTATE may overcome logistical disadvantages of68 Ga-based tracers and facilitate SSTR-targeted PET/CT imaging of thyroid carcinoma., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
22. Opposing Effects of Cannabidiol in Patient-derived Neuroendocrine Tumor, Pheochromocytoma/Paraganglioma Primary Cultures.
- Author
-
Wang K, Schober L, Fischer A, Bechmann N, Maurer J, Peischer L, Reul A, Hantel C, Reincke M, Beuschlein F, Robledo M, Mohr H, Pellegata NS, Schilbach K, Knösel T, Ilmer M, Angele M, Kroiss M, Maccio U, Broglie-Däppen M, Vetter D, Lehmann K, Pacak K, Grossman AB, Auernhammer CJ, Zitzmann K, and Nölting S
- Subjects
- Humans, Female, Male, Middle Aged, Adult, Tumor Cells, Cultured, Aged, Young Adult, Cell Line, Tumor, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Primary Cell Culture, Cannabidiol pharmacology, Paraganglioma drug therapy, Paraganglioma pathology, Pheochromocytoma drug therapy, Pheochromocytoma pathology, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors pathology, Adrenal Gland Neoplasms drug therapy, Adrenal Gland Neoplasms pathology
- Abstract
Context: Treatment options for advanced neuroendocrine tumors (NETs), pheochromocytomas and paragangliomas (PPGLs) are still limited. In recent years, antitumor effects of cannabinoids have been reported; however, there are only very limited data available in NETs or PPGLs., Objective: Investigation of the effects of cannabidiol (CBD) on patient-derived human NET/PPGL primary cultures and on NET/PPGL cell lines., Methods: We established primary cultures derived from 46 different patients with PPGLs (n = 35) or NETs (n = 11) who underwent tumor resection at 2 centers. Treatment of patient primary cultures with clinically relevant doses (5 µM) and slightly higher doses (10 µM) of CBD was performed., Results: We found opposing effects of 5 µM CBD: significant antitumor effects in 5/35 (14%) and significant tumor-promoting effects in 6/35 (17%) of PPGL primary cultures. In terms of antitumor effects, cluster 2-related PPGLs showed significantly stronger responsivity to CBD compared to cluster 1-related PPGLs (P = .042). Of the cluster 2-related tumors, NF1 PPGLs showed the strongest responsivity (4/5 PPGL primary cultures with a significant decrease in cell viability were NF1-mutated). We also found opposing effects of 10 µM CBD in PPGLs and NETs: significant antitumor effects in 9/33 of PPGL (27%) and 3/11 of NET (27%) primary cultures and significant tumor-promoting effects in 6/33 of PPGL (18%) and 2/11 of NET (18%) primary cultures., Conclusion: We suggest a potential novel treatment option for some NETs/PPGLs but also provide evidence for caution when applying cannabinoids as supportive therapy for pain or appetite management to cancer patients and possibly as health supplements., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
23. Peptide-guided adaptor-CAR T-Cell therapy for the treatment of SSTR2-expressing neuroendocrine tumors.
- Author
-
Pellegrino C, Favalli N, Volta L, Benz R, Puglioli S, Bassi G, Zitzmann K, Auernhammer CJ, Nölting S, Magnani CF, Neri D, Beuschlein F, and Manz MG
- Subjects
- Animals, Humans, Mice, Xenograft Model Antitumor Assays, Cell Line, Tumor, T-Lymphocytes immunology, T-Lymphocytes metabolism, Receptors, Somatostatin genetics, Receptors, Somatostatin metabolism, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen metabolism, Immunotherapy, Adoptive methods, Neuroendocrine Tumors therapy, Neuroendocrine Tumors immunology, Neuroendocrine Tumors pathology
- Abstract
Somatostatin receptor type 2 (SSTR2) is one of the five subtypes of somatostatin receptors and is overexpressed on the surface of most gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs), pituitary tumors, paraganglioma, and meningioma, as well as hepatocellular carcinoma and breast cancer. Chimeric antigen receptor (CAR) T-cells are genetically engineered to express an artificial, T-cell activating binder, leading upon ligation to biocidal activity against target-antigen expressing cells. Adaptor-CAR T-cells recognize, via the CAR, a tag on an antigen-binding molecule, building an activating bridge between the CAR and the target cell. We hypothesized that a novel fluorescent-peptide antagonist of SSTR2, called Octo-Fluo, in combination with anti-FITC adaptor CAR (AdFITC(E2)-CAR) T-cells, may function as an on-off tunable activating bridge between the CAR and SSTR2 expressing target cells. In vitro studies confirmed the binding of Octo-Fluo to Bon1-SSTR2 mCherry-Luc cells without evidence of internalization. AdFITC(E2)-CAR T-cells were activated and efficiently induced Bon1-SSTR2 cell death in vitro, in an Octo-Fluo concentration-dependent manner. Similarly, AdFITC(E2)-CAR T-cells in combination with Octo-Fluo efficiently infiltrated the tumor and eliminated Bon1-SSTR2 tumors in immunodeficient mice in therapeutic settings. Both, AdFITC(E2)-CAR T-cell tumor infiltration and biocidal activity were Octo-Fluo concentration-dependent, with high doses of Octo-Fluo, saturating both the CAR and the SSTR2 antigen independently, leading to the loss of tumor infiltration and biocidal activity due to the loss of bridge formation. Our findings demonstrate the potential of using AdFITC(E2)-CAR T-cells with Octo-Fluo as a versatile, on-off tunable bispecific adaptor for targeted CAR T-cell immunotherapy against SSTR2-positive NETs., Competing Interests: CP, GB, NF, DN, FB and MGM are inventors of a patent application that describes the Octo-Fluo molecule., (© 2024 The Author(s). Published with license by Taylor & Francis Group, LLC.)
- Published
- 2024
- Full Text
- View/download PDF
24. [Imaging of neuroendocrine tumors of the gastrointestinal tract : Value of (hybrid) imaging diagnostics in radiology].
- Author
-
Ebner R, Rübenthaler J, Ricke J, Sheikh GT, Unterrainer LM, Auernhammer CJ, Spitzweg C, Brendel M, Schmid-Tannwald C, and Cyran CC
- Subjects
- Humans, Magnetic Resonance Imaging methods, Multimodal Imaging methods, Positron Emission Tomography Computed Tomography methods, Positron-Emission Tomography methods, Tomography, X-Ray Computed methods, Gastrointestinal Neoplasms diagnostic imaging, Gastrointestinal Neoplasms pathology, Gastrointestinal Neoplasms diagnosis, Neuroendocrine Tumors diagnostic imaging, Neuroendocrine Tumors pathology
- Abstract
Clinical/methodical Issue: Neuroendocrine tumors (NET) represent a heterogeneous group of rare tumors that predominantly arise in the gastrointestinal tract. At the time of initial diagnosis, the NET has already spread locoregionally in about half of the patients, and 27% of patients have already developed distant metastases. Since this plays a crucial role in therapy planning, accurate diagnostic imaging is important., Standard Radiological Methods: Due to its high temporal and spatial resolution (multiphasic including arterial phase), computed tomography (CT) plays a decisive role in primary staging and follow-up care, while magnetic resonance imaging (MRI) with its excellent soft tissue contrast offers advantages in the assessment of parenchymal organs in the upper abdomen., Methodical Innovations: Somatostatin receptor (SSR) positron emission tomography (PET) provides additional functional information that not only helps to detect the primary tumor and distant metastases, but also has a significant influence on therapeutic management in a theranostic approach., Performance: Hybrid imaging using SSR-PET/CT has proven to be particularly effective in the detection of NET. Compared to conventional imaging, it provides additional information in 68% of patients, which has a significant impact on clinical management., Achievements: Imaging of NET requires the combined use of various methods such as ultrasound, CT, MRI, and PET/CT to enable accurate diagnosis and effective treatment planning., Practical Recommendations: SSR-PET/CT is a valuable tool for the accurate staging of neuroendocrine tumors of the gastrointestinal tract, especially with small metastases, while MRI with hepatocyte-specific contrast agent and diffusion-weighted imaging is useful for the specific assessment of liver metastases., (© 2024. The Author(s), under exclusive licence to Springer Medizin Verlag GmbH, ein Teil von Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
25. [Imaging of pancreatic neuroendocrine tumors].
- Author
-
Berger F, Ingenerf M, Auernhammer CJ, Cyran C, Ebner R, Zacherl M, Ricke J, and Schmid-Tannwald C
- Subjects
- Humans, Multimodal Imaging methods, Positron Emission Tomography Computed Tomography methods, Tomography, X-Ray Computed methods, Magnetic Resonance Imaging methods, Neuroendocrine Tumors diagnostic imaging, Neuroendocrine Tumors pathology, Neuroendocrine Tumors secondary, Pancreatic Neoplasms diagnostic imaging, Pancreatic Neoplasms pathology
- Abstract
Background: Neuroendocrine tumors of the pancreas have a broad biological spectrum. The treatment decision is based on an optimal diagnosis with regard to the local findings and possible locoregional and distant metastases. In addition to purely morphologic imaging procedures, functional parameters are playing an increasingly important role in imaging., Objectives: Prerequisites for optimal imaging of the pancreas, technical principles are provided, and the advantages and disadvantages of common cross-sectional imaging techniques as well as clinical indications for these special imaging methods are discussed., Materials and Methods: Guidelines, basic and review papers will be analyzed., Results: Neuroendocrine tumors of the pancreas have a broad imaging spectrum. Therefore, there is a need for multimodality imaging in which morphologic and functional techniques support each other. While positron emission tomography/computed tomography (PET/CT) can determine the presence of one or more lesions and its/their functional status of the tumor, magnetic resonance imaging (MRI) efficiently identifies the location, relationship to the main duct and the presence of liver metastases. CT allows a better vascular evaluation, even in the presence of anatomical variants as well as sensitive detection of lung metastases., Conclusions: Knowledge of the optimal combination of imaging modalities including clinical and histopathologic results and dedicated imaging techniques is essential to achieve an accurate diagnosis to optimize treatment decision-making and to assess therapy response., (© 2024. The Author(s), under exclusive licence to Springer Medizin Verlag GmbH, ein Teil von Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
26. Neuroendocrine liver metastases treated using transarterial radioembolization: Identification of prognostic parameters at 68Ga-DOTATATE PET/CT.
- Author
-
Ingenerf M, Grawe F, Winkelmann M, Karim H, Ruebenthaler J, Fabritius MP, Ricke J, Seidensticker R, Auernhammer CJ, Zacherl MJ, Seidensticker M, and Schmid-Tannwald C
- Subjects
- Male, Humans, Middle Aged, Positron Emission Tomography Computed Tomography methods, Prognosis, Ki-67 Antigen, Gallium Radioisotopes, Positron-Emission Tomography, Liver Neoplasms diagnostic imaging, Liver Neoplasms therapy, Liver Neoplasms secondary, Organometallic Compounds, Neuroendocrine Tumors diagnostic imaging, Neuroendocrine Tumors therapy, Neuroendocrine Tumors secondary
- Abstract
Purpose: To identify prognostic clinical and imaging parameters for patients with neuroendocrine liver metastases (NELMs) undergoing transarterial radioembolization (TARE)., Materials and Methods: Forty-seven patients (27 men; mean age, 64 years) with NELMs who received TARE, along with pre-procedure liver MRI and
68 Ga-DOTATATE positron emission tomography/computed tomography were included. Apparent diffusion coefficient and standardized uptake value (SUV) of three liver metastases, normal spleen and liver were measured. SUVmax or SUVmean were used for the calculation of tumor-to-organ ratios (tumor-to-spleen and tumor-to-liver ratios) using all possible combinations (including SUVmax/SUVmax, SUVmax/SUVmean, and SUVmean/SUVmean). Clinical parameters (hepatic tumor-burden, presence of extra-hepatic metastases, chromograninA, Ki-67 and bilirubin levels) were assessed. Overall survival, progression-free survival (PFS) and hepatic progression-free survival (HPFS) were calculated using Kaplan-Meier curves., Results: Median overall survival, PFS and HPFS were 49.6, 13.1 and 28.3 months, respectively. In multivariable Cox regression analysis, low Ki-67 (≤ 5%), low hepatic tumor-burden (< 10%), absence of extrahepatic metastases, and increased Tmean/Lmax ratio were significant prognostic factors of longer overall survival and HPFS. High baseline chromograninA (> 1330 ng/mL) was associated with shorter HPFS. Tmean/Lmax > 1.9 yielded a median overall survival of 69 vs. 33 months (P < 0.04), and a median HPFS of 30 vs. 19 months (P = 0.09). For PFS, high baseline SUVmax of NELMs was the single significant parameter in the multivariable model. SUVmax > 28 resulted in a median PFS of 16.9 vs. 6.5 months, respectively (P = 0.001)., Conclusion: High preinterventional Tmean/Lmax ratios, and high SUVmax on68 Ga-DOTATATE positron emission tomography/computed tomography seem to have prognostic value in patients with NELMs undergoing TARE, potentially aiding patient selection and management alongside conventional variables., Competing Interests: Declaration of Competing Interest The authors have no conflicts of interest related to this work to declare., (Copyright © 2023 Société française de radiologie. Published by Elsevier Masson SAS. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
27. Impact of the PI3K-alpha inhibitor alpelisib on everolimus resistance and somatostatin receptor expression in an orthotopic pancreatic NEC xenograft mouse model.
- Author
-
Mohan AM, Prasad S, Schmitz-Peiffer F, Lange C, Lukas M, Koziolek EJ, Albrecht J, Messroghli D, Stein U, Ilmer M, Wang K, Schober L, Reul A, Maurer J, Friemel J, Weber A, Zuellig RA, Hantel C, Fritsch R, Reincke M, Pacak K, Grossman AB, Auernhammer CJ, Beuschlein F, Brenner W, Beindorff N, and Nölting S
- Subjects
- Humans, Female, Animals, Mice, Everolimus pharmacology, Everolimus therapeutic use, Receptors, Somatostatin, Phosphatidylinositol 3-Kinases, Heterografts, Positron Emission Tomography Computed Tomography, Mice, SCID, Phosphoinositide-3 Kinase Inhibitors therapeutic use, Neuroendocrine Tumors pathology, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms pathology
- Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) inhibitor everolimus is one of the few approved therapies for locally advanced and metastatic neuroendocrine tumours (NETs). However, after initial disease stabilisation, most patients develop resistance within 1 year. Our aim was to overcome resistance to everolimus by additional treatment with the PI3K-alpha inhibitor alpelisib in an everolimus-resistant orthotopic pancreatic neuroendocrine carcinoma xenograft mouse model. Female SCID mice underwent laparoscopic pancreatic transplantation of everolimus-sensitive (BON1KDMSO) or everolimus-resistant (BON1RR2) NET cells. Both groups were further divided into four treatment groups: placebo, everolimus, alpelisib, and everolimus + alpelisib (combination). Oral treatment was started at a tumour volume of approximately 140 mm3 and continued until 1900-2000 mm3, validated by weekly MRI. Somatostatin receptor expression and tumour viability were analysed by 68Ga-DOTATOC and 18F-FDG PET/CT. Everolimus resistance of the BON1RR2 tumours was confirmed. In the everolimus-sensitive group, everolimus alone, alpelisib alone, and combination treatment significantly prolonged survival, compared to placebo, while in the BON1RR2 group, only combination treatment significantly prolonged survival compared to placebo, but neither everolimus nor alpelisib alone. Placebo-treated everolimus-sensitive tumours grew more rapidly (median survival 45 days), compared to placebo-treated everolimus-resistant tumours (60 days). Within the everolimus-sensitive group, the combination-treated mice showed the longest median survival (52 days). Of all groups, the everolimus-resistant combination-treated group survived longest (69 days). Combination treatment with everolimus and alpelisib seems promising to overcome everolimus resistance in neuroendocrine neoplasms, and should be further examined in a clinical trial.
- Published
- 2023
- Full Text
- View/download PDF
28. Responses to systemic therapy in metastatic pheochromocytoma/paraganglioma: a retrospective multicenter cohort study.
- Author
-
Fischer A, Kloos S, Remde H, Dischinger U, Pamporaki C, Timmers HJLM, Robledo M, Fliedner SMJ, Wang K, Maurer J, Reul A, Bechmann N, Hantel C, Mohr H, Pellegata NS, Bornstein SR, Kroiss M, Auernhammer CJ, Reincke M, Pacak K, Grossman AB, Beuschlein F, and Nölting S
- Subjects
- Humans, Iodine Radioisotopes therapeutic use, Retrospective Studies, Cohort Studies, Temozolomide therapeutic use, Sunitinib therapeutic use, Succinate Dehydrogenase genetics, Somatostatin therapeutic use, Pheochromocytoma pathology, Paraganglioma genetics, Adrenal Gland Neoplasms pathology, Brain Neoplasms, Neoplasms, Second Primary, Cardiovascular Diseases
- Abstract
Objective: The therapeutic options for metastatic pheochromocytomas/paragangliomas (mPPGLs) include chemotherapy with cyclophosphamide/vincristine/dacarbazine (CVD), temozolomide monotherapy, radionuclide therapies, and tyrosine kinase inhibitors such as sunitinib. The objective of this multicenter retrospective study was to evaluate and compare the responses of mPPGLs including those with pathogenic variants in succinate dehydrogenase subunit B (SDHB), to different systemic treatments., Design: This is a retrospective analysis of treatment responses of mPPGL patients (n = 74) to systemic therapies., Methods: Patients with mPPGLs treated at 6 specialized national centers were selected based on participation in the ENSAT registry. Survival until detected progression (SDP) and disease-control rates (DCRs) at 3 months were evaluated based on imaging reports., Results: For the group of patients with progressive disease at baseline (83.8% of 74 patients), the DCR with first-line CVD chemotherapy was 75.0% (n = 4, SDP 11 months; SDHB [n = 1]: DCR 100%, SDP 30 months), with somatostatin peptide receptor-based radionuclide therapy (PPRT) 85.7% (n = 21, SDP 17 months; SDHB [n = 10]: DCR 100%, SDP 14 months), with 131I-meta-iodobenzylguanidine (131I-MIBG) 82.6% (n = 23, SDP 43 months; SDHB [n = 4]: DCR 100%, SDP 24 months), with sunitinib 100% (n = 7, SDP 18 months; SDHB [n = 3]: DCR 100%, SDP 18 months), and with somatostatin analogs 100% (n = 4, SDP not reached). The DCR with temozolomide as second-line therapy was 60.0% (n = 5, SDP 10 months; SDHB [n = 4]: DCR 75%, SDP 10 months)., Conclusions: We demonstrate in a real-life clinical setting that all current therapies show reasonable efficacy in preventing disease progression, and this is equally true for patients with germline SDHB mutations., Competing Interests: Conflict of interest: None declared., (© The Author(s) 2023. Published by Oxford University Press on behalf of European Society of Endocrinology.)
- Published
- 2023
- Full Text
- View/download PDF
29. PD-L1 and HIF-2α Upregulation in Head and Neck Paragangliomas after Embolization.
- Author
-
Fischer A, Maccio U, Wang K, Friemel J, Broglie Daeppen MA, Vetter D, Lehmann K, Reul A, Robledo M, Hantel C, Bechmann N, Pacak K, Zitzmann K, Auernhammer CJ, Grossman AB, Beuschlein F, and Nölting S
- Abstract
Hypoxia activates pathways associated with tumor progression, metastatic spread, and alterations in the immune microenvironment leading to an immunosuppressive phenotype. In particular, the upregulation of PD-L1, a target for therapy with checkpoint inhibitors, is well-studied in several tumors. However, the relationship between hypoxia and PD-L1 regulation in pheochromocytomas and paragangliomas (PPGL), and especially in paragangliomas treated with embolization, is still largely unexplored. We investigated the expression of the hypoxia-marker HIF-2α and of PD-L1 in a PPGL-cohort with and without embolization as potential biomarkers that may predict the response to treatment with HIF-2α and checkpoint inhibitors. A total of 29 tumor samples from 25 patients who were operated at a single center were included and analyzed utilizing immunohistochemistry (IHC) for PD-L1 and HIF-2α. Embolization prior to surgery was performed in seven (24%) tumors. PD-L1 expression in tumor cells of head and neck paragangliomas (HNPGLs) receiving prior embolization (median PD-L1 positivity: 15%) was significantly higher as compared to PD-L1 expression in HNPGLs without prior embolization (median PD-L1 positivity: 0%) ( p = 0.008). Consistently, significantly more HNPGLs with prior embolization were positive for HIF-2α (median nuclear HIF-2α positivity: 40%) as compared to HNPGLs without prior embolization (median nuclear HIF-2α positivity: 0%) ( p = 0.016). Our results support the hypothesis that embolization with subsequent hypoxia leads to the upregulation of both PD-L1 and HIF-2α in HNPGLs, and could thus facilitate targeted treatment with HIF-2α and checkpoint inhibitors in the case of inoperable, locally advanced, or metastatic disease.
- Published
- 2023
- Full Text
- View/download PDF
30. Diagnostic accuracy of SSR-PET/CT compared to histopathology in the identification of liver metastases from well-differentiated neuroendocrine tumors.
- Author
-
Fabritius MP, Soltani V, Cyran CC, Ricke J, Bartenstein P, Auernhammer CJ, Spitzweg C, Schnitzer ML, Ebner R, Mansournia S, Hinterberger A, Lohse A, Sheikh GT, Winkelmann M, Knösel T, Ingenerf M, Schmid-Tannwald C, Kunz WG, Rübenthaler J, and Grawe F
- Subjects
- Humans, Positron Emission Tomography Computed Tomography, Receptors, Somatostatin, Positron-Emission Tomography methods, Fluorodeoxyglucose F18, Sensitivity and Specificity, Radiopharmaceuticals, Neuroendocrine Tumors diagnostic imaging, Neuroendocrine Tumors pathology, Liver Neoplasms diagnostic imaging
- Abstract
Background: Histopathology is the reference standard for diagnosing liver metastases of neuroendocrine tumors (NETs). Somatostatin receptor-positron emission tomography / computed tomography (SSR-PET/CT) has emerged as a promising non-invasive imaging modality for staging NETs. We aimed to assess the diagnostic accuracy of SSR-PET/CT in the identification of liver metastases in patients with proven NETs compared to histopathology., Methods: Histopathologic reports of 139 resected or biopsied liver lesions of patients with known NET were correlated with matching SSR-PET/CTs and the positive/negative predictive value (PPV/NPV), sensitivity, specificity, and diagnostic accuracy of SSR-PET/CT were evaluated. PET/CT reading was performed by one expert reader blinded to histopathology and clinical data., Results: 133 of 139 (95.7%) liver lesions showed malignant SSR-uptake in PET/CT while initial histopathology reported on 'liver metastases of NET´ in 127 (91.4%) cases, giving a PPV of 91.0%. Re-biopsy of the initially histopathologically negative lesions (reference standard) nevertheless diagnosed 'liver metastases of NET' in 6 cases, improving the PPV of PET/CT to 95.5%. Reasons for initial false-negative histopathology were inadequate sampling in the sense of non-target biopsies. The 6 (4.3%) SSR-negative lesions were all G2 NETs with a Ki-67 between 2-15%., Conclusion: SSR-PET/CT is a highly accurate imaging modality for the diagnosis of liver metastases in patients with proven NETs. However, we found that due to the well-known tumor heterogeneity of NETs, specifically in G2 NETs approximately 4-5% are SSR-negative and may require additional imaging with [
18 F]FDG PET/CT., (© 2023. International Cancer Imaging Society (ICIS).)- Published
- 2023
- Full Text
- View/download PDF
31. Validation of the SSTR-RADS 1.0 for the structured interpretation of SSTR-PET/CT and treatment planning in neuroendocrine tumor (NET) patients.
- Author
-
Grawe F, Ebner R, Geyer T, Beyer L, Winkelmann M, Sheikh GT, Eschbach R, Schmid-Tannwald C, Cyran CC, Ricke J, Bartenstein P, Heimer MM, Faggioni L, Spitzweg C, Fabritius MP, Auernhammer CJ, and Ruebenthaler J
- Subjects
- Humans, Receptors, Somatostatin, Reproducibility of Results, Radionuclide Imaging, Positron Emission Tomography Computed Tomography, Neuroendocrine Tumors diagnostic imaging, Neuroendocrine Tumors therapy, Neuroendocrine Tumors pathology
- Abstract
Objectives: The recently proposed standardized reporting and data system for somatostatin receptor (SSTR)-targeted PET/CT SSTR-RADS 1.0 showed promising first results in the assessment of diagnosis and treatment planning with peptide receptor radionuclide therapy (PRRT) in neuroendocrine tumors (NET). This study aimed to determine the intra- and interreader agreement of SSTR-RADS 1.0., Methods: SSTR-PET/CT scans of 100 patients were independently evaluated by 4 readers with different levels of expertise according to the SSTR-RADS 1.0 criteria at 2 time points within 6 weeks. For each scan, a maximum of five target lesions were freely chosen by each reader (not more than three lesions per organ) and stratified according to the SSTR-RADS 1.0 criteria. Overall scan score and binary decision on PRRT were assessed. Intra- and interreader agreement was determined using the intraclass correlation coefficient (ICC)., Results: Interreader agreement using SSTR-RADS 1.0 for identical target lesions (ICC ≥ 0.91) and overall scan score (ICC ≥ 0.93) was excellent. The decision to state "functional imaging fulfills requirements for PRRT and qualifies patient as potential candidate for PRRT" also demonstrated excellent agreement among all readers (ICC ≥ 0.86). Intrareader agreement was excellent even among different experience levels when comparing target lesion-based scores (ICC ≥ 0.98), overall scan score (ICC ≥ 0.93), and decision for PRRT (ICC ≥ 0.88)., Conclusion: SSTR-RADS 1.0 represents a highly reproducible and accurate system for stratifying SSTR-targeted PET/CT scans with high intra- and interreader agreement. The system is a promising approach to standardize the diagnosis and treatment planning in NET patients., Key Points: • SSTR-RADS 1.0 offers high reproducibility and accuracy. • SSTR-RADS 1.0 is a promising method to standardize diagnosis and treatment planning for patients with NET., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
32. Immuno-histochemical correlation of fibrosis-related markers with the desmoplastic reaction of the mesentery in small intestine neuroendocrine neoplasms.
- Author
-
Bösch F, Altendorf-Hofmann A, Koliogiannis V, Ilhan H, Jacob S, Pretzsch E, Nölting S, Werner J, Klauschen F, Auernhammer CJ, Angele MK, and Knösel T
- Subjects
- Humans, Fibrosis, Intestine, Small pathology, Mesentery pathology, Intestinal Neoplasms pathology, Neuroendocrine Tumors pathology
- Abstract
Introduction: Small intestine neuroendocrine neoplasms (siNENs) will attain more importance due to their increasing incidence. Moreover, siNENs might lead to a desmoplastic reaction (DR) of the mesentery causing severe complications and deteriorating prognosis. The expression of fibrosis-related proteins appears to be the key mechanisms for the development of this desmoplastic reaction. Therefore, this study aimed to investigate the association of the desmoplastic mesentery with specific fibrosis-related protein expression levels., Materials and Methods: By immunohistochemistry, the protein expression levels of four fibrosis-related markers (APLP2, BNIP3L, CD59, DKK3) were investigated in primary tumors of 128 siNENs. The expression levels were correlated with the presence of a desmoplastic reaction and clinico-pathological parameters., Results: In the primary tumor, APLP2, BNIP3L, CD59 and DKK3 were highly expressed in 29.7% (n = 38), 64.9% (n = 83), 92.2% (n = 118) and 80.5% (n = 103), respectively. There was no significant correlation of a single marker or the complete marker panel to the manifestation of a desmoplastic mesentery. The desmoplastic mesentery was significantly associated with clinical symptoms, such as flushing and diarrhea. However, neither the fibrosis-related marker panel nor single marker expressions were associated with clinical symptoms., Discussion: The expression rates of four fibrosis-related markers in the primary tumor display a distinct pattern. However, the expression patterns are not associated with desmoplastic altered mesenteric lymph node metastases and the expression patterns did not correlate with prognosis. These findings suggest alternative mechanisms being responsible for the desmoplastic reaction., (© 2022. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
33. Diagnostic performance of PET/CT in the detection of liver metastases in well-differentiated NETs.
- Author
-
Grawe F, Rosenberger N, Ingenerf M, Beyer L, Eschbach R, Todica A, Seidensticker R, Schmid-Tannwald C, Cyran CC, Ricke J, Bartenstein P, Auernhammer CJ, Ruebenthaler J, and Fabritius MP
- Subjects
- Humans, Positron Emission Tomography Computed Tomography methods, Positron-Emission Tomography methods, Retrospective Studies, Magnetic Resonance Imaging methods, Sensitivity and Specificity, Liver Neoplasms pathology, Neuroendocrine Tumors diagnostic imaging, Neuroendocrine Tumors pathology
- Abstract
Background: The aim of this retrospective study was to compare the diagnostic accuracy of somatostatin receptor (SSR)-PET/CT to liver MRI as reference standard in the evaluation of hepatic involvement in neuroendocrine tumors (NET)., Methods: An institutional database was screened for "SSR" imaging studies between 2006 and 2021. 1000 NET Patients (grade 1/2) with 2383 SSR-PET/CT studies and matching liver MRI in an interval of +3 months were identified. Medical reports of SSR-PET/CT and MRI were retrospectively evaluated regarding hepatic involvement and either confirmed by both or observed in MRI but not in SSR-PET/CT (false-negative) or in SSR-PET but not in MRI (false-positive)., Results: Metastatic hepatic involvement was reported in 1650 (69.2%) of the total 2383 SSR-PET/CT imaging studies, whereas MRI detected hepatic involvement in 1685 (70.7%) cases. There were 51 (2.1%) false-negative and 16 (0.7%) false-positive cases. In case of discrepant reports, MRI and PET/CT were reviewed side by side for consensus reading. SSR-PET/CT demonstrated a sensitivity of 97.0% (95%CI: 96.0%, 97.7%), a specificity of 97.7% (95%CI: 96.3%, 98.7%), a PPV of 99.0% (95%CI: 98.4%, 99.4%) and NPV of 93.0% (95%CI: 91.0, 94.8%) in identifying hepatic involvement. The most frequent reason for false-negative results was the small size of lesions with the majority < 0.6 cm., Conclusion: This study confirms the high diagnostic accuracy of SSR-PET/CT in the detection of hepatic involvement in NET patients based on a patient-based analysis of metastatic hepatic involvement with a high sensitivity and specificity using liver MRI imaging as reference standard. However, one should be aware of possible pitfalls when a single imaging method is used in evaluating neuroendocrine liver metastases in patients., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
34. Comparison of somatostatin receptor expression in patients with neuroendocrine tumours with and without somatostatin analogue treatment imaged with [ 18 F]SiTATE.
- Author
-
Eschbach RS, Hofmann M, Späth L, Sheikh GT, Delker A, Lindner S, Jurkschat K, Wängler C, Wängler B, Schirrmacher R, Tiling R, Brendel M, Wenter V, Dekorsy FJ, Zacherl MJ, Todica A, Ilhan H, Grawe F, Cyran CC, Unterrainer M, Rübenthaler J, Knösel T, Paul T, Boeck S, Westphalen CB, Spitzweg C, Auernhammer CJ, Bartenstein P, Unterrainer LM, and Beyer L
- Abstract
Purpose: Somatostatin analogues (SSA) are frequently used in the treatment of neuroendocrine tumours. Recently, [
18 F]SiTATE entered the field of somatostatin receptor (SSR) positron emission tomography (PET)/computed tomography (CT) imaging. The purpose of this study was to compare the SSR-expression of differentiated gastroentero-pancreatic neuroendocrine tumours (GEP-NET) measured by [18F]SiTATE-PET/CT in patients with and without previous treatment with long-acting SSAs to evaluate if SSA treatment needs to be paused prior to [18F]SiTATE-PET/CT., Methods: 77 patients were examined with standardised [18F]SiTATE-PET/CT within clinical routine: 40 patients with long-acting SSAs up to 28 days prior to PET/CT examination and 37 patients without pre-treatment with SSAs. Maximum and mean standardized uptake values (SUVmax and SUVmean) of tumours and metastases (liver, lymphnode, mesenteric/peritoneal and bones) as well as representative background tissues (liver, spleen, adrenal gland, blood pool, small intestine, lung, bone) were measured, SUV ratios (SUVR) were calculated between tumours/metastases and liver, likewise between tumours/metastases and corresponding specific background, and compared between the two groups., Results: SUVmean of liver (5.4 ± 1.5 vs. 6.8 ± 1.8) and spleen (17.5 ± 6.8 vs. 36.7 ± 10.3) were significantly lower (p < 0.001) and SUVmean of blood pool (1.7 ± 0.6 vs. 1.3 ± 0.3) was significantly higher (p < 0.001) in patients with SSA pre-treatment compared to patients without. No significant differences between tumour-to-liver and specific tumour-to-background SUVRs were observed between both groups (all p > 0.05)., Conclusion: In patients previously treated with SSAs, a significantly lower SSR expression ([18F]SiTATE uptake) in normal liver and spleen tissue was observed, as previously reported for 68Ga-labelled SSAs, without significant reduction of tumour-to-background contrast. Therefore, there is no evidence that SSA treatment needs to be paused prior to [18F]SiTATE-PET/CT., Competing Interests: CA has received research contracts Novartis, lecture honorarium Ipsen, Novartis, Advanced Accelerator Applications and honoraria for advisory boards Advanced Accelerator Applications. HI has received research contracts Novartis. LB received honoraria for advisory boards Bayer, Advanced Accelerator Applications and is a Novartis Radiopharmaceuticals GmbH employee since 10/2022. CW has received honoraria from Amgen, Bayer, Chugai, Celgene, Falk, GSK, MSD, Merck, Janssen, Ipsen, Roche, Servier, SIRTeX, Taiho; served on advisory boards for Bayer, BMS, Celgene, Servier, Shire/Baxalta, Rafael Pharmaceuticals, RedHill, Roche, has received travel support by Bayer, Celgene, RedHill, Roche, Servier, Taiho and research grants institutional by Roche. CW serves as an officer for European Society of Medical Oncology ESMO, Deutsche Krebshilfe DKH, Arbeitsgemeinschaft internistische Onkologie AIO. The remaining authors declared that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Eschbach, Hofmann, Späth, Sheikh, Delker, Lindner, Jurkschat, Wängler, Wängler, Schirrmacher, Tiling, Brendel, Wenter, Dekorsy, Zacherl, Todica, Ilhan, Grawe, Cyran, Unterrainer, Rübenthaler, Knösel, Paul, Boeck, Westphalen, Spitzweg, Auernhammer, Bartenstein, Unterrainer and Beyer.)- Published
- 2023
- Full Text
- View/download PDF
35. Treatment Assessment of pNET and NELM after Everolimus by Quantitative MRI Parameters.
- Author
-
Ingenerf M, Kiesl S, Winkelmann M, Auernhammer CJ, Rübenthaler J, Grawe F, Fabritius MP, Ricke J, and Schmid-Tannwald C
- Abstract
Assessment of treatment response to targeted therapies such as everolimus is difficult, especially in slow-growing tumors such as NETs. In this retrospective study, 17 patients with pancreatic neuroendocrine tumors (pNETs) and hepatic metastases (NELMs) (42 target lesions) who received everolimus were analyzed. Intralesional signal intensities (SI) of non-contrast T1w, T2w and DCE imaging, and apparent diffusion coefficients (ADCmean and ADCmin) of DWI, were measured on baseline and first follow-up MRI after everolimus initiation. Response assessment was categorized according to progression-free survival (PFS), with responders (R) showing a PFS of ≥11 months. ADCmin of NELMs decreased in Rs whereas it increased in non-responders (NR). Percentual changes of ADCmin and ADCmean differed significantly between response groups (p < 0.03). By contrast, ADC of the pNETs tended to increase in Rs, while there was no change in NRs. Tumor-to-liver (T/L) ratio of T1 SI of NELMs increased in Rs and decreased in NRs, and percentual changes differed significantly between response groups (p < 0.02). T1 SI of the pNETs tended to decrease in Rs and increase in Ns. The quotient of pretherapeutic and posttherapeutic ADCmin values (DADCmin) and length of everolimus treatment showed significant association with PFS in univariable Cox analysis. In conclusion, quantitative MRI, especially DWI, seems to allow treatment assessment of pNETs with NELMs under everolimus. Interestingly, the responding NELMs showed decreasing ADC values, and there might be an opposite effect on ADC and T1 SI between NELMs and pNETs.
- Published
- 2022
- Full Text
- View/download PDF
36. Impact of tumor size and location on endoscopic ultrasound-guided sampling of pancreatic neuroendocrine tumors: A recursive partitioning analysis.
- Author
-
Sirtl S, Mahajan UM, Auernhammer CJ, Dziadkiewicz P, Hohmann E, Wójcik M, Kos-Kudła B, Hartleb M, Knösel T, Schirra J, Mayerle J, Schulz C, and Żorniak M
- Subjects
- Endoscopic Ultrasound-Guided Fine Needle Aspiration, Humans, Retrospective Studies, Neuroectodermal Tumors, Primitive, Neuroendocrine Tumors pathology, Pancreatic Neoplasms pathology
- Abstract
Background: Current guidelines provide weak recommendations to treat small (<2 cm) non-functional pancreatic neuroendocrine tumors with low Ki-67 proliferation index either by resection or clinical follow-up. However, there is a lack of consensus regarding the minimal size of pNET, which allows EUS-guided biopsy with high enough diagnostic accuracy for stratification., Methods: We conducted a retrospective, bicentric analysis of patients who had undergone EUS-guided pNET sampling in two tertiary care Endoscopy Units in Germany and Poland. Using a recursive partitioning of the tree-aided model, we aimed to stratify the probability of successful EUS-guided biopsy of pNET lesions according to their size and location., Results: In our pNET cohort, successful histological confirmation of a pNET diagnosis was achieved in 59/69 (85.5%) cases at the initial EUS-guided biopsy. In 41 patients with a pNET size less than 18.5 mm, the EUS-guided first biopsy was successful in 90.2%. In 16 of these patients with smaller lesions, EUS-guided sampling was 100% in very small (less than 11 mm) and extremely small lesions (less than 8 mm). The biopsy success rate was 100% in tail lesions in the size range between ≥5.95 and <8.1 mm but only 33.3% independent of the investigator in pancreatic head or body, with an error rate of 11.2% CONCLUSION: Using a recursive partitioning of the tree-aided stratification model, we demonstrate for the first time that in balancing risks and benefits, very small pNETs (<1 cm) in the tail of the pancreas should be sampled under EUS-guidance., Competing Interests: Declaration of competing interest The authors declare no potential conflicts of interest concerning the research, authorship, and/or publication of this article., (Copyright © 2022 IAP and EPC. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
37. Personalized drug testing in human pheochromocytoma/paraganglioma primary cultures.
- Author
-
Wang K, Schütze I, Gulde S, Bechmann N, Richter S, Helm J, Lauseker M, Maurer J, Reul A, Spoettl G, Klink B, William D, Knösel T, Friemel J, Bihl M, Weber A, Fankhauser M, Schober L, Vetter D, Broglie Däppen M, Ziegler CG, Ullrich M, Pietzsch J, Bornstein SR, Lottspeich C, Kroiss M, Fassnacht M, Wenter VUJ, Ladurner R, Hantel C, Reincke M, Eisenhofer G, Grossman AB, Pacak K, Beuschlein F, Auernhammer CJ, Pellegata NS, and Nölting S
- Subjects
- Animals, Everolimus therapeutic use, Humans, Mice, Zoledronic Acid therapeutic use, Adrenal Gland Neoplasms drug therapy, Adrenal Gland Neoplasms genetics, Adrenal Gland Neoplasms metabolism, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Paraganglioma drug therapy, Paraganglioma genetics, Paraganglioma pathology, Pheochromocytoma drug therapy, Pheochromocytoma genetics, Pheochromocytoma metabolism
- Abstract
Aggressive pheochromocytomas and paragangliomas (PPGLs) are difficult to treat, and molecular targeting is being increasingly considered, but with variable results. This study investigates established and novel molecular-targeted drugs and chemotherapeutic agents for the treatment of PPGLs in human primary cultures and murine cell line spheroids. In PPGLs from 33 patients, including 7 metastatic PPGLs, we identified germline or somatic driver mutations in 79% of cases, allowing us to assess potential differences in drug responsivity between pseudohypoxia-associated cluster 1-related (n = 10) and kinase signaling-associated cluster 2-related (n = 14) PPGL primary cultures. Single anti-cancer drugs were either more effective in cluster 1 (cabozantinib, selpercatinib, and 5-FU) or similarly effective in both clusters (everolimus, sunitinib, alpelisib, trametinib, niraparib, entinostat, gemcitabine, AR-A014418, and high-dose zoledronic acid). High-dose estrogen and low-dose zoledronic acid were the only single substances more effective in cluster 2. Neither cluster 1- nor cluster 2-related patient primary cultures responded to HIF-2a inhibitors, temozolomide, dabrafenib, or octreotide. We showed particular efficacy of targeted combination treatments (cabozantinib/everolimus, alpelisib/everolimus, alpelisib/trametinib) in both clusters, with higher efficacy of some targeted combinations in cluster 2 and overall synergistic effects (cabozantinib/everolimus, alpelisib/trametinib) or synergistic effects in cluster 2 (alpelisib/everolimus). Cabozantinib/everolimus combination therapy, gemcitabine, and high-dose zoledronic acid appear to be promising treatment options with particularly high efficacy in SDHB-mutant and metastatic tumors. In conclusion, only minor differences regarding drug responsivity were found between cluster 1 and cluster 2: some single anti-cancer drugs were more effective in cluster 1 and some targeted combination treatments were more effective in cluster 2.
- Published
- 2022
- Full Text
- View/download PDF
38. Development of severe intrapulmonary shunting in a patient with carcinoid heart disease after closure of a persistent foramen ovale: a case report.
- Author
-
Schüttler D, Mourouzis K, Auernhammer CJ, and Rizas KD
- Abstract
Background: Neuroendocrine tumours (NETs) can affect the cardiopulmonary system causing carcinoid heart disease (CHD) and valve destruction. Persistent foramen ovale (PFO) occlusion is indicated in patients with CHD and shunt-related left heart valve involvement., Case Summary: We report the case of a 54-year-old female patient with metastatic NET originating from the small bowel. The patient was on medication with octreotide and telotristat. One year after diagnosis, cardiac involvement of carcinoid developed with regurgitation of right-sided and, due to PFO, left-sided heart valves. Closure of PFO was performed (Occlutech 16/18 mm). One year later, she presented with recurrent severe dyspnoea. The PFO occluder was in situ without residual shunt. Valvular heart disease, including left-sided disease, and metastatic spread of NET were stable. Blood gas analysis revealed arterial hypoxaemia (pO
2 = 44 mmHg/5.87 kPa), which was related to extensive intrapulmonary shunting (31% shunt fraction) confirmed using contrast-enhanced echocardiography. The patient was prescribed long-term oxygen supplementation as symptomatic therapy and anti-tumoural therapy was intensified with selective internal radiotherapy (SIRT) of the liver metastases to improve biochemical control of the carcinoid syndrome. At a follow-up visit 4 months after SIRT, the patient-reported stable dyspnoea; however, magnetic resonance imaging revealed progression of osseous metastases., Discussion: An echocardiographic assessment of the presence of a PFO is recommended in patients with NET as PFO closure minimizes the risk of left-sided carcinoid valve disease. Deterioration of symptomatic status in metastasized NET might also be due to a hepatopulmonary-like physiology with intrapulmonary shunting and arterial desaturation thought to be caused by vasoactive substances secreted by the tumour. This is a rare case describing the development of this syndrome after PFO closure., (© The Author(s) 2021. Published by Oxford University Press on behalf of the European Society of Cardiology.)- Published
- 2021
- Full Text
- View/download PDF
39. Dosimetry and optimal scan time of [ 18 F]SiTATE-PET/CT in patients with neuroendocrine tumours.
- Author
-
Beyer L, Gosewisch A, Lindner S, Völter F, Mittlmeier LM, Tiling R, Brendel M, Cyran CC, Unterrainer M, Rübenthaler J, Auernhammer CJ, Spitzweg C, Böning G, Gildehaus FJ, Jurkschat K, Wängler C, Wängler B, Schirrmacher R, Wenter V, Todica A, Bartenstein P, and Ilhan H
- Subjects
- Adult, Computers, Female, Humans, Male, Positron-Emission Tomography, Radiometry, Tissue Distribution, Neuroendocrine Tumors diagnostic imaging, Positron Emission Tomography Computed Tomography
- Abstract
Purpose: Radiolabelled somatostatin analogues targeting somatostatin receptors (SSR) are well established for combined positron emission tomography/computer tomography (PET/CT) imaging of neuroendocrine tumours (NET). [
18 F]SiTATE has recently been introduced showing high image quality, promising clinical performance and improved logistics compared to the clinical reference standard68 Ga-DOTA-TOC. Here we present the first dosimetry and optimal scan time analysis., Methods: Eight NET patients received a [18 F]SiTATE-PET/CT (250 ± 66 MBq) with repeated emission scans (10, 30, 60, 120, 180 min after injection). Biodistribution in normal organs and SSR-positive tumour uptake were assessed. Dosimetry estimates for risk organs were determined using a combined linear-monoexponential model, and by applying18 F S-values and reference target masses for the ICRP89 adult male or female (OLINDA 2.0). Tumour-to-background ratios were compared quantitatively and visually between different scan times., Results: After 1 h, normal organs showed similar tracer uptake with only negligible changes until 3 h post-injection. In contrast, tracer uptake by tumours increased progressively for almost all types of metastases, thus increasing tumour-to-background ratios over time. Dosimetry resulted in a total effective dose of 0.015 ± 0.004 mSv/MBq. Visual evaluation revealed no clinically relevant discrepancies between later scan times, but image quality was rated highest in 60 and 120 min images., Conclusion: [18 F]SiTATE-PET/CT in NET shows overall high tumour-to-background ratios from 60 to 180 min after injection and an effective dose comparable to68 Ga-labelled alternatives. For clinical use of [18 F]SiTATE, the best compromise between image quality and tumour-to-background contrast is reached at 120 min, followed by 60 min after injection., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
40. Treatment of advanced gastroenteropancreatic neuroendocrine neoplasia, are we on the way to personalised medicine?
- Author
-
Rinke A, Auernhammer CJ, Bodei L, Kidd M, Krug S, Lawlor R, Marinoni I, Perren A, Scarpa A, Sorbye H, Pavel ME, Weber MM, Modlin I, and Gress TM
- Subjects
- Biomarkers, Tumor, Epigenesis, Genetic, Humans, Intestinal Neoplasms diagnosis, Intestinal Neoplasms genetics, Neuroendocrine Tumors diagnosis, Neuroendocrine Tumors genetics, Pancreatic Neoplasms diagnosis, Pancreatic Neoplasms genetics, Stomach Neoplasms diagnosis, Stomach Neoplasms genetics, Intestinal Neoplasms therapy, Neuroendocrine Tumors therapy, Pancreatic Neoplasms therapy, Precision Medicine methods, Stomach Neoplasms therapy
- Abstract
Gastroenteropancreatic neuroendocrine neoplasia (GEPNEN) comprises clinically as well as prognostically diverse tumour entities often diagnosed at late stage. Current classification provides a uniform terminology and a Ki67-based grading system, thereby facilitating management. Advances in the study of genomic and epigenetic landscapes have amplified knowledge of tumour biology and enhanced identification of prognostic and potentially predictive treatment subgroups. Translation of this genomic and mechanistic biology into advanced GEPNEN management is limited. 'Targeted' treatments such as somatostatin analogues, peptide receptor radiotherapy, tyrosine kinase inhibitors and mammalian target of rapamycin inhibitors are treatment options but predictive tools are lacking. The inability to identify clonal heterogeneity and define critical oncoregulatory pathways prior to therapy, restrict therapeutic efficacy as does the inability to monitor disease status in real time. Chemotherapy in the poor prognosis NEN G3 group, though associated with acceptable response rates, only leads to short-term tumour control and their molecular biology requires delineation to provide new and more specific treatment options.The future requires an exploration of the NEN tumour genome, its microenvironment and an identification of critical oncologic checkpoints for precise drug targeting. In the advance to personalised medical treatment of patients with GEPNEN, clinical trials need to be based on mechanistic and multidimensional characterisation of each tumour in order to identify the therapeutic agent effective for the individual tumour.This review surveys advances in NEN research and delineates the current status of translation with a view to laying the basis for a genome-based personalised medicine management of advanced GEPNEN., Competing Interests: Competing interests: AR has received honoraria for presentations and advisory boards from AAA, Advanz Pharma, Falk, IPSEN and Novartis. CJA has received research contracts (Ipsen, Novartis), lecture honorarium (AAA, Ipsen, Novartis) and advisory board honorarium (Novartis). LB has been non remunerated consultant or speaker for AAA-Novartis, Ipsen, Curium, Clovis, ITM, Iba and has received research support from AAA-Novartis. MK is Laboratory Director for Wren Laboratories LLC. HS received research grants from Novartis, Amgen and Ipsen, honoraria for advisory board for Novartis, Pfizer, Keocyt, AstraZeneca and Hutchinson, honoraria for presentations from Novartis, BMS, Roche, Amgen, Ipsen, Merck, Shire, Celgene and Bayer. TMG has received funding from IPSEN, Pfizer and Novartis for joined research projects, participation in advisory boards and lectures. IMM is medical and scientific consultant for Clifton Life Sciences. MEP received honoraria for participation in advisory boards and presentations from Novartis, IPSEN, Pfizer Riemser and Lexicon, honoraria for presentations from Prime Oncology and research funding from IPSEN and Novartis. MMW received honoraria for participation in advisory boards and presentations from IPSEN, AAA/Novartis and Advanz Pharma and research funding from Merck., (© Author(s) (or their employer(s)) 2021. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
41. Molecular Imaging with 18 F-FDG PET/CT and 99m Tc-MIBI SPECT/CT in Osteitis Fibrosa Cystica Generalisata.
- Author
-
Holzgreve A, Fabritius MP, Knösel T, Mittlmeier LM, Rübenthaler J, Tiling R, Auernhammer CJ, Bartenstein P, and Unterrainer M
- Abstract
Benign so-called "brown tumors" secondary to hyperparathyroidism are a rare diagnostic pitfall due to their impressively malignant-like character in various imaging modalities. We present the case of a 65-year-old male patient with multiple unclear osteolytic lesions on prior imaging suspicious for metastatic malignant disease. Eventually, findings of
18 F-FDG PET/CT staging and99m Tc-MIBI scintigraphy resulted in revision of the initially suspected malignant diagnosis. This case illustrates how molecular imaging findings non-invasively corroborate the correct diagnosis of osteitis fibrosa cystica generalisata with the formation of multiple benign brown tumors.- Published
- 2021
- Full Text
- View/download PDF
42. Feasibility and Early Clinical Experience of Online Adaptive MR-Guided Radiotherapy of Liver Tumors.
- Author
-
Rogowski P, von Bestenbostel R, Walter F, Straub K, Nierer L, Kurz C, Landry G, Reiner M, Auernhammer CJ, Belka C, Niyazi M, and Corradini S
- Abstract
Purpose: To assess the feasibility and early results of online adaptive MR-guided radiotherapy (oMRgRT) of liver tumors., Methods: We retrospectively examined consecutive patients with primary or secondary liver lesions treated at our institution using a 0.35T hybrid MR-Linac (Viewray Inc., Mountain View, CA, USA). Online-adaptive treatment planning was used to account for interfractional anatomical changes, and real-time intrafractional motion management using online 2D cine MRI was performed using a respiratory gating approach. Treatment response and toxicity were assessed during follow-up., Results: Eleven patients and a total of 15 lesions were evaluated. Histologies included cholangiocarcinomas and metastases of neuroendocrine tumors, colorectal carcinomas, sarcomas and a gastrointestinal stroma tumor. The median BED
10 of the PTV prescription doses was 84.4 Gy (range 59.5-112.5 Gy) applied in 3-5 fractions and the mean GTV BED10 was in median 147.9 Gy (range 71.7-200.5 Gy). Online plan adaptation was performed in 98% of fractions. The median overall treatment duration was 53 min. The treatment was feasible and successfully completed in all patients. After a median follow-up of five months, no local failure occurred and no ≥ grade two toxicity was observed. OMRgRT resulted in better PTV coverage and fewer OAR constraint violations., Conclusion: Early results of MR-linac based oMRgRT for the primary and secondary liver tumors are promising. The treatment was feasible in all cases and well tolerated with minimal toxicity. The technique should be compared to conventional SBRT in further studies to assess the advantages of the technique.- Published
- 2021
- Full Text
- View/download PDF
43. Antitumoral Activity of the MEK Inhibitor Trametinib (TMT212) Alone and in Combination with the CDK4/6 Inhibitor Ribociclib (LEE011) in Neuroendocrine Tumor Cells In Vitro.
- Author
-
Jin XF, Spöttl G, Maurer J, Nölting S, and Auernhammer CJ
- Abstract
Objectives: This study assessed the antitumoral activity of the MEK inhibitor trametinib (TMT212) and the ERK1/2 inhibitor SCH772984, alone and in combination with the CDK4/6 inhibitor ribociclib (LEE011) in human neuroendocrine tumor (NET) cell lines in vitro., Methods: Human NET cell lines BON1, QGP-1, and NCI-H727 were treated with trametinib or SCH772984, alone and in combination with ribociclib, to assess cell proliferation, cell cycle distribution, and protein signaling using cell proliferation, flow cytometry, and Western blot assays, respectively., Results: Trametinib and SCH772984, alone and in combination with ribociclib, significantly reduced NET cell viability and arrested NET cells at the G1 phase of the cell cycle in all three cell lines tested. In addition, trametinib also caused subG1 events and apoptotic PARP cleavage in QGP1 and NCI-H727 cells. A western blot analysis demonstrated the use of trametinib alone and trametinib in combination with ribociclib to decrease the expression of pERK, cMyc, Chk1, pChk2, pCDK1, CyclinD1, and c-myc in a time-dependent manner in NCI-H727 and QGP-1 cells., Conclusions: MEK and ERK inhibition causes antiproliferative effects in human NET cell lines in vitro. The combination of the MEK inhibitor trametinib (TMT212) with the CDK4/6 inhibitor ribociclib (LEE011) causes additive antiproliferative effects. Future preclinical and clinical studies of MEK inhibition in NETs should be performed.
- Published
- 2021
- Full Text
- View/download PDF
44. Cost-Effectiveness Analysis of 68 Ga DOTA-TATE PET/CT, 111 In-Pentetreotide SPECT/CT and CT for Diagnostic Workup of Neuroendocrine Tumors.
- Author
-
Froelich MF, Schnitzer ML, Holzgreve A, Gassert FG, Gresser E, Overhoff D, Schwarze V, Fabritius MP, Nörenberg D, von Münchhausen N, Hokamp NG, Auernhammer CJ, Ilhan H, Todica A, and Rübenthaler J
- Abstract
Neuroendocrine tumors (NETs) are relatively rare neoplasms arising from the hormone-producing neuroendocrine system that can occur in various organs such as pancreas, small bowel, stomach and lung. As the majority of these tumors express somatostatin receptors (SSR) on their cell membrane, utilization of SSR analogs in nuclear medicine is a promising, but relatively costly approach for detection and localization. The aim of this study was to analyze the cost-effectiveness of
68 Ga-DOTA-TATE PET/CT (Gallium-68 DOTA-TATE Positron emission tomography/computed tomography) compared to111 In-pentetreotide SPECT/CT (Indium-111 pentetreotide Single Photon emission computed tomography/computed tomography) and to CT (computed tomography) alone in detection of NETs. A decision model on the basis of Markov simulations evaluated lifetime costs and quality-adjusted life years (QALYs) related to either a CT, SPECT/CT or PET/CT. Model input parameters were obtained from publicized research projects. The analysis is grounded on the US healthcare system. Deterministic sensitivity analysis of diagnostic parameters and probabilistic sensitivity analysis predicated on a Monte Carlo simulation with 30,000 reiterations was executed. The willingness-to-pay (WTP) was determined to be $ 100,000/QALY. In the base-case investigation, PET/CT ended up with total costs of $88,003.07 with an efficacy of 4.179, whereas CT ended up with total costs of $88,894.71 with an efficacy of 4.165. SPECT/CT ended up with total costs of $89,973.34 with an efficacy of 4.158. Therefore, the strategies CT and SPECT/CT were dominated by PET/CT in the base-case scenario. In the sensitivity analyses, PET/CT remained a cost-effective strategy. This result was due to reduced therapy costs of timely detection. The additional costs of68 Ga-DOTA-TATE PET/CT when compared to CT alone are justified in the light of potential savings in therapy costs and better outcomes.- Published
- 2021
- Full Text
- View/download PDF
45. Mixed large cell neuroendocrine carcinoma and squamous cell carcinoma of the colon: detailed molecular characterisation of two cases indicates a distinct colorectal cancer entity.
- Author
-
Woischke C, Jung P, Jung A, Kumbrink J, Eisenlohr S, Auernhammer CJ, Vieth M, Kirchner T, and Neumann J
- Subjects
- Biomarkers, Tumor analysis, Carcinoma, Large Cell chemistry, Carcinoma, Large Cell pathology, Carcinoma, Large Cell therapy, Carcinoma, Neuroendocrine chemistry, Carcinoma, Neuroendocrine pathology, Carcinoma, Neuroendocrine therapy, Carcinoma, Squamous Cell chemistry, Carcinoma, Squamous Cell pathology, Carcinoma, Squamous Cell therapy, Colonic Neoplasms chemistry, Colonic Neoplasms pathology, Colonic Neoplasms therapy, Fatal Outcome, Female, Genetic Predisposition to Disease, Humans, Male, Middle Aged, Neoplasms, Complex and Mixed chemistry, Neoplasms, Complex and Mixed pathology, Neoplasms, Complex and Mixed therapy, Phenotype, Treatment Outcome, Biomarkers, Tumor genetics, Carcinoma, Large Cell genetics, Carcinoma, Neuroendocrine genetics, Carcinoma, Squamous Cell genetics, Colonic Neoplasms genetics, F-Box-WD Repeat-Containing Protein 7 genetics, Mutation, Neoplasms, Complex and Mixed genetics
- Abstract
We present two rare cases of mixed large cell neuroendocrine carcinoma and squamous cell carcinoma of the colon. A literature search revealed only three published cases with similar histology but none of these reports provided profound molecular and mutational analyses. Our two cases exhibited a distinct, colon-like immunophenotype with strong nuclear CDX2 and β-catenin expression in more than 90% of the tumour cells of both components. We analysed the two carcinomas regarding microsatellite stability, RAS, BRAF and PD-L1 status. In addition, next-generation panel sequencing with Ion AmpliSeq™ Cancer Hotspot Panel v2 was performed. This approach revealed mutations in FBXW7, CTNNB1 and PIK3CA in the first case and FBXW7 and RB1 mutations in the second case. We looked for similar mutational patterns in three publicly available colorectal adenocarcinoma data sets, as well as in collections of colorectal mixed neuroendocrine-non-neuroendocrine neoplasms (MiNENs) and colorectal neuroendocrine carcinomas. This approach indicated that the FBXW7 point mutation, without being accompanied by classical adenoma-carcinoma sequence mutations, such as APC, KRAS and TP53, likely occurs at a relatively high frequency in mixed neuroendocrine and squamous cell carcinoma and therefore may be characteristic for this rare tumour type. FBXW7 codifies the substrate recognition element of an ubiquitin ligase, and inactivating FBXW7 mutations lead to an exceptional accumulation of its target β-catenin which results in overactivation of the Wnt-signalling pathway. In line with previously described hypotheses of de-differentiation of colon cells by enhanced Wnt-signalling, our data indicate a crucial role for mutant FBXW7 in the unusual morphological switch that determines these rare neoplasms. Therefore, mixed large cell neuroendocrine and a squamous cell carcinoma can be considered as a distinct carcinoma entity in the colon, defined by morphology, immunophenotype and distinct molecular genetic alteration(s)., (© 2020 The Authors. The Journal of Pathology: Clinical Research published by The Pathological Society of Great Britain and Ireland & John Wiley & Sons, Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
46. Distinct Expression Patterns of VEGFR 1-3 in Gastroenteropancreatic Neuroendocrine Neoplasms: Supporting Clinical Relevance, but not a Prognostic Factor.
- Author
-
Bösch F, Altendorf-Hofmann A, Jacob S, Auernhammer CJ, Spitzweg C, Boeck S, Schubert-Fritschle G, Werner J, Kirchner T, Angele MK, and Knösel T
- Abstract
Introduction: Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are an increasing tumor entity. Since many patients are diagnosed at an advanced stage, treatment is still challenging and dependent on many tumor and patient specific factors. Therefore, the aim of the present study was to elucidate the expression rates and the prognostic value of vascular endothelial growth factor receptor (VEGFR) 1-3 in GEP-NENs. A potential association to immune checkpoint markers was further investigated., Material and Methods: The expression levels of VEGFR 1-3 were analyzed by immunohistochemistry and correlated with the expression of the checkpoint markers PD-1 and PD-L1. Furthermore, the tumor samples of 249 GEP-NEN patients were studied and correlated with overall survival rates and clinicopathological features. Kaplan-Meier analyses and the log rank test were used for survival analyses. Categorical variables were compared by the χ2 test., Results: The most common primary tumor site was the small intestine (50.6%), followed by the pancreas (25.7%). VEGFR 1 was highly expressed in 59%, VEGFR 2 in 6.4%, and VEGFR 3 in 61.8% of the analyzed samples. The expression of VEGFR 1-3 was not significantly associated with survival rates. Pancreatic NENs had the highest expression of VEGFR 1 and 3 in 80% of the cases. VEGFR 1-3 positivity correlated with the expression levels of immune checkpoint markers., Discussion: VEGFR 1-3 show a distinct expression pattern in different subgroups of neuroendocrine neoplasias indicating a conceivable target. Moreover, there was a substantial association between VEGFRs and immune checkpoint markers. Taken together, anti-VEGFR therapy represents a promising therapeutic approach in GEP-NEN patients and should be addressed in future studies.
- Published
- 2020
- Full Text
- View/download PDF
47. Radioembolization for neuroendocrine liver metastases is safe and effective prior to major hepatic resection.
- Author
-
Bösch F, Ilhan H, Pfahler V, Thomas M, Knösel T, Eibl V, Pratschke S, Bartenstein P, Seidensticker M, Auernhammer CJ, Spitzweg C, Guba MO, Werner J, and Angele MK
- Abstract
Background: Radioembolization (RE) is well established in the treatment of neuroendocrine liver metastases. However surgery is rarely performed after RE, although liver resection is the gold standard in the treatment of localized neuroendocrine liver metastases. Therefore, aim of the present study was to evaluate the safety and feasibility of liver resection after RE in a homogenous cohort., Methods: From a prospective surgical (n=494) and nuclear medical (n=138) database patients with NELM who underwent liver resection and/or RE were evaluated. Between September 2011 and December 2017 eight patients could be identified who underwent liver resection after RE (mean therapeutic activity of 1,746 Mbq). Overall and progression free survival were evaluated as well as epidemiological and perioperative factors. The surgical specimens were analyzed for necrosis, fibrosis, inflammation, and steatosis., Results: The mean hepatic tumor load of patients, who had liver surgery after RE, was 31.4% with a mean Ki-67 proliferation index of 5.9%. The majority of these patients (7/8) received whole liver RE prior to liver resection, which did not increase morbidity and mortality compared to a surgical collective. Indications for RE were oncological (6/8) or carcinoid syndrome associated reasons (2/8). Mean overall survival was 25.1 months after RE and subsequent surgery. Tumor necrosis in radioembolized lesions was 29.4% without evidence of fibrosis and inflammation in hepatic tissue., Conclusions: This is the first study analyzing the multimodal therapeutic approach of liver resection following whole liver RE. This treatment algorithm is safe, does not lead to an increased morbidity and is associated with a favorable oncological outcome. Nonetheless, patient selection remains a key issue., Competing Interests: Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/hbsn.2019.07.11). HI reports speaker honoraria from SIRTEX Medical. CJA has received research contracts (Novartis, Ipsen, ITM Solucin), lecture honorarium (Novartis, Ipsen, Falk Foundation) and advisory board honorarium (Novartis). CS reports personal fees from Ipsen, personal fees from Novartis, personal fees from Shire, personal fees from Blueprint Medicine, personal fees from Bayer, personal fees from Eisai, outside the submitted work. MS reports grants and personal fees from SIRTEX Medical, grants and personal fees from BAYER, personal fees from Siemens, personal fees from Cook, personal fees from Boston Scientific, outside the submitted work. The other authors have no conflicts of interest to declare., (2020 Hepatobiliary Surgery and Nutrition. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
48. Biodistribution and first clinical results of 18 F-SiFAlin-TATE PET: a novel 18 F-labeled somatostatin analog for imaging of neuroendocrine tumors.
- Author
-
Ilhan H, Lindner S, Todica A, Cyran CC, Tiling R, Auernhammer CJ, Spitzweg C, Boeck S, Unterrainer M, Gildehaus FJ, Böning G, Jurkschat K, Wängler C, Wängler B, Schirrmacher R, and Bartenstein P
- Subjects
- Humans, Positron Emission Tomography Computed Tomography, Positron-Emission Tomography, Receptors, Somatostatin metabolism, Retrospective Studies, Somatostatin, Tissue Distribution, Neuroendocrine Tumors diagnostic imaging, Organometallic Compounds
- Abstract
Introduction: PET/CT using
68 Ga-labeled somatostatin analogs (SSA) targeting somatostatin receptors (SSR) on the cell surface of well-differentiated neuroendocrine tumors (NET) represents the clinical reference standard for imaging. However, economic and logistic challenges of the68 Ge/68 Ga generator-based approach have disadvantages over18 F-labeled compounds. Here, we present the first in-human data of18 F-SiFAlin-TATE, a novel18 F-labeled, SSR-targeting peptide. The aim was to compare the intra-individual biodistribution, tumor uptake, and image quality of18 F-SiFAlin-TATE to the clinical reference standard68 Ga-DOTA-TOC., Methods: Thirteen patients with NET staged with both68 Ga-DOTA-TOC and18 F-SiFAlin-TATE PET/CT have been included in this retrospective analysis. We compared the biodistribution in normal organs and tumor uptake of NET lesions by SUVmean and SUVmax measurement for tracers. Additionally mean and max tumor-to-liver (TLR) and tumor-to-spleen ratios (TSR) have been calculated by division of SUVmean and SUVmax of tumor lesions by the SUVmean of the liver and spleen, respectively. Additionally, image quality was visually rated by 5 blinded readers and an intra-class correlation (ICC) analysis on inter-observer agreement has been performed., Results: Compared with68 Ga-DOTA-TOC, the biodistribution of18 F-SiFAlin-TATE showed somewhat higher, however, statistically not significant higher uptake in the liver, spleen, and adrenal glands. Significantly higher uptake was observed in the kidneys. Tumor uptake was higher in most tumor lesions with significantly higher uptake in common metastatic sites of NET including the liver (SUVmax 18.8 ± 8.4 vs. 12.8 ± 5.6; p < 0.001), lymph nodes (SUVmax 23.8 ± 20.7 vs. 17.4 ± 16.1; p < 0.001) and bone (SUVmax 16.0 ± 10.1 vs. 10.3 ± 5.7; p < 0.01) for18 F-SiFAlin-TATE. The high tumor uptake resulted in favorable TLR and TSR, comparable with that of68 Ga-DOTA-TOC. The ICC analysis on the inter-observer agreement on image quality was substantial and almost perfect. Image quality was rated as excellent in most cases in both68 Ga-DOTA-TOC and18 F-SiFAlin-TATE PET., Conclusion: The favorable characteristics of18 F-SiFAlin-TATE with a high image quality, the kit-like labeling procedure, and the promising clinical performance enable improved logistics and diagnostic possibilities for PET imaging of NET. Our first clinical results warrant further systematic studies investigating the clinical use of18 F-SiFAlin-TATE in NET patients.- Published
- 2020
- Full Text
- View/download PDF
49. Inhibition of Wnt/β-Catenin Signaling in Neuroendocrine Tumors in vitro: Antitumoral Effects.
- Author
-
Jin XF, Spoettl G, Maurer J, Nölting S, and Auernhammer CJ
- Abstract
Background and Aims: Inhibition of Wnt/β-catenin signaling by specific inhibitors is currently being investigated as an antitumoral strategy for various cancers. The role of Wnt/β-catenin signaling in neuroendocrine tumors still needs to be further investigated., Methods: This study investigated the antitumor activity of the porcupine (PORCN) inhibitor WNT974 and the β-catenin inhibitor PRI-724 in human neuroendocrine tumor (NET) cell lines BON1, QGP-1, and NCI-H727 in vitro. NET cells were treated with WNT974, PRI-724, or small interfering ribonucleic acids against β-catenin, and subsequent analyses included cell viability assays, flow cytometric cell cycle analysis, caspase3/7 assays and Western blot analysis., Results: Treatment of NET cells with WNT974 significantly reduced NET cell viability in a dose- and time-dependent manner by inducing NET cell cycle arrest at the G1 and G2/M phases without inducing apoptosis. WNT974 primarily blocked Wnt/β-catenin signaling by the dose- and time-dependent downregulation of low-density lipoprotein receptor-related protein 6 (LRP6) phosphorylation and non-phosphorylated β-catenin and total β-catenin, as well as the genes targeting the latter (c-Myc and cyclinD1). Furthermore, the WNT974-induced reduction of NET cell viability occurred through the inhibition of GSK-3-dependent or independent signaling (including pAKT/mTOR, pEGFR and pIGFR signaling). Similarly, treatment of NET cells with the β-catenin inhibitor PRI-724 caused significant growth inhibition, while the knockdown of β-catenin expression by siRNA reduced NET tumor cell viability of BON1 cells but not of NCI-H727 cells., Conclusions: The PORCN inhibitor WNT974 possesses antitumor properties in NET cell lines by inhibiting Wnt and related signaling. In addition, the β-catenin inhibitor PRI-724 possesses antitumor properties in NET cell lines. Future studies are needed to determine the role of Wnt/β-catenin signaling in NET as a potential therapeutic target., Competing Interests: This study has been funded by an unrestricted research grant to C.J.A. from Novartis Pharma GmbH, Nuernberg, Germany. C.J.A. has received research contracts (Ipsen, Novartis, ITM Solucin), lecture honorarium (Ipsen, Novartis, Falk Foundation) and advisory board honorarium (Novartis). S.N. has received research contracts (Novartis) and lecture fees (Ipsen). The other authors declare no conflicts of interest in this work.
- Published
- 2020
- Full Text
- View/download PDF
50. [The Thyroid and Pregnancy].
- Author
-
Koehler VF, Auernhammer CJ, and Spitzweg C
- Subjects
- Female, Humans, Practice Guidelines as Topic, Pregnancy, Thyroid Function Tests, Pregnancy Complications, Thyroid Diseases, Thyroid Gland physiology
- Abstract
Pregnancy has a profound impact on the thyroid gland and its function. This has to be considered in the assessment of thyroid function tests on the basis of trimester-specific reference intervals during pregnancy and in the decision making when to start therapy.The adverse impact of overt thyroid disorders in pregnancy is well understood, while the relevance of subclinical thyroid disorders and presence of thyroid antibodies remains a bit controversial. In euthyroid pregnant women with positive thyroid antibodies, levothyroxine (LT4) therapy may be discussed individually in the case of recurrent abortions. Furthermore, the risk of adverse pregnancy outcomes seems to be increased in the presence of both thyroid-stimulating hormone (TSH)-elevation and positive thyroid antibodies. Therefore, in case of subclinical hypothyroidism, taking in consideration the thyroid peroxidase antibody (TPO-Ab)-status individual decision-making and liberal initiation of LT4 therapy is recommended. In contrast, overt hypothyroidism is a strong indication for LT4 administration, aiming at rapid achievement of euthyroidism.The most common cause of hyperthyroidism is transient gestational thyrotoxicosis mediated by human chorionic gonadotropin (hCG), which leads to a reduction or suppression of TSH in the first trimester that does not require antithyroid medication. In other causes of overt hyperthyroidism antithyroid drugs (propylthiouracil or thionamides) need to be considered carefully and require interdisciplinary management.The presented recommendations are based on the current guideline of the American Thyroid Association (ATA) and the European Thyroid Association (ETA) as well as recently published literature., Competing Interests: Die Autoren geben an, dass kein Interessenkonflikt besteht., (© Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.