TY - THES A1 - Willmes, Christoph T1 - Therapie kutaner Tumoren : Identifizierung molekularer Biomarker der ex vivo Chemosensitivität des malignen Melanoms und Evaluierung der Wirkungsweise von Interferonen und Artemisininen auf das Merkelzellkarzinom T1 - Treatment of cutaneous tumors N2 - Für Patienten mit malignem Melanom im Stadium der Fernmetastasierung gibt es bis heute lediglich Therapieoptionen mit sehr eingeschränkten Erfolgsaussichten. Diese Tatsache bestätigt die Notwendigkeit von Biomarkern zur Vorhersage des Erfolgs verschiedener Therapien. Der ATP-basierende ex vivo Chemosensitivitätsassay hat sich als erfolgreiche Methode zur individuellen Vorhersage eines Chemotherapieerfolgs herausgestellt. Tatsächlich zeigte der Assay ein heterogenes Sensitivitätsprofil gegen verschiedene Chemotherapeutika und ließ in getesteten Patienten ein ex vivo wirksames Chemotherapieregime identifizieren, das anschließend auch klinische Therapieerfolge bei Verwendung der Therapie mit dem besten individuellen Chemosensitivitätsindex(BICSI) zeigte. Um diesen sehr aufwendigen Assay zukünftig zu umgehen, sollten in der vorliegenden Arbeit prädiktive molekulare Biomarker der Chemosensitivität identifiziert werden. Hierfür wurden im Voraus durch einen Microarray die Kandidaten Secernin 1 (SCRN1), Lysyl oxidaselike 1 (LOXL1), Thymosin beta 4 X-linked (TMSB4X), Vesicle-associated membrane protein 5 (VAMP5) und Serine protease inhibitor B1 (SERPINB1) als differentiell exprimierte Gene in chemosensitivem gegenüber chemoresistentem Gewebe identifiziert. Die relative Expression dieser Kandidatengene wurde daraufhin in bis zu 128 verschiedenen Melanomgeweben mit dem Chemosensitivitätsindex verschiedener Chemotherapeutika korreliert. Hierbei konnte eine signifikante Korrelation zwischen SerpinB1 mit der Chemosensitivität gegenüber der Therapiekombination mit Paclitaxel und Cisplatin auf Gen- aber nicht auf Proteinebene identifiziert werden. Weiterhin konnte eine differentielle Expression ebenfalls in chemosensitiven und -resistenten Melanomzelllinien nachgewiesen werden, die allerdings im Vergleich mit dem analysierten Gewebe in gegensätzlicher Richtung verlief. Zusammenfassend lässt sich sagen, dass SerpinB1 ein vielversprechender Marker für die Chemosensitivität gegenüber Paclitaxel und Cisplatin ist, dessen funktionelle Bedeutung aber unklar bleibt. Das Merkelzellkarzinom (MCC) ist ein seltener und hoch aggressiver Tumor der mit dem Merkelzellpolyomavirus (MCV) in Zusammenhang steht. Da MCC Zelllinien zur Aufrechterhaltung ihrer Viabilität die MCV T-Antigene benötigen, könnte der Einsatz von Interferonen (IFN) ein möglicher therapeutischer Ansatz zur Behandlung dieser Krebserkrankung sein. In der vorliegenden Arbeit haben wir daher die Effekte von IFNs auf MCC Zelllinien, mit besonderer Berücksichtigung der MCV+ Linien, untersucht. IFNs vom Typ I (hier Multiferon, ein Mix verschiedener IFN α Subtypen, und IFN β) wirkten stark inhibierend auf die zelluläre Viabilität. Die Zellzyklusanalyse zeigte eine Erhöhung des sub-G Anteils der Zellen nach Behandlung mit IFN, was auf Apoptose als ausschlagebenden Grund schließen ließ. Diese Effekte waren für die Behandlung mit IFN β weniger stark ausgeprägt. Der inhibitorische Effekt von Typ I IFNs auf MCV+ MCC Zelllinien war assoziiert mit einer verringerten Expression des viralen großen T-Antigens (LTA) und einer Erhöhung in der Expression von promyelocytic leukemia protein (PML), das dafür bekannt ist, die Funktion des LTA störend zu beeinflussen. Zusätzlich führte die intratumorale Anwendung von Multiferon in vivo zu einer Regression im Wachstum von MCV+, aber nicht MCV- MCC Xenotransplantaten. Die Ergebnisse zeigen das Typ I IFNs einen starken antitumoralen Effekt haben, der zum Teil durch die Regulierung des LTA herbeigeführt wird. Neben diesen direkten Effekten der IFNs auf die Zellproliferation induzieren diese auch die Expression von MHC Klasse I Molekülen in MCC Zelllinien. Die Durchflusszytometrie zeigte eine Induktion der MHC Klasse I Expression in drei MHC I negativen MCC Zelllinien und eine Erhöhung der Expression, die vor der Behandlung eine geringe Menge an MHC I aufwiesen. Diese Effekte konnten auch in den in vivo Xenotransplantaten beobachtet werden. Die Ergebnisse zeigen, dass die Behandlung mit IFN sowohl direkte als auch indirekte Effekte auf das MCC hat und eine breite Anwendung in Patienten mit MCV+ und MCV- Tumoren finden kann. Neben IFNs sind auch Artemisinin und seine Derivate bekannt für ihre antitumoralen und antiviralen Eigenschaften. Daher haben wir den Effekt des Artemisininderivats Artesunate auf MCV+ und MCV- MCC Zelllinien getestet. Tatsächlich konnten wir auch hier einen antiproliferativen Effekt des Stoffes nachweisen, der stärker auf MCV+ als auf MCV- Zelllinien wirkte und bei ersteren wiederum mit einer reduzierten LTA Expression einherging. Im Vergleich dazu blieben Fibroblasten von der Behandlung unbeeinflusst. Das verringerte Tumorwachstum konnte ebenfalls für in vivo Xenotransplantationsmodelle gezeigt werden. Auf Grundlage dieser Erkenntnis sollte eine genauere Untersuchung dieses alten Naturheilstoffes für die Behandlung von MCC Patienten in Betracht gezogen werden. N2 - For melanoma patients with distant metastases all available therapeutic options demonstrate only very limited efficacy up to date. This fact substantiates the need of predictive markers for therapy response. For example, ex-vivo chemosensitivity testing by an ATP-based luminescence assay is a promising tool to predict the individual outcome of different chemotherapy regimens. Indeed, this assay demonstrates a heterogeneous chemosensitivity against different cytotoxic drugs which correlates with chemotherapy outcome in terms of therapy response and overall survival; for the treatment of the patient the drug with the best individual chemosensitivity index(BICSI) is used. To circumvent this elaborate assay in the future, we want to identify and characterize predictive molecular biomarkers of specific chemosensitivity. Initially, predictive biomarker aspirants were identified by a microarray comparing chemosensitive and chemoresistant melanoma cell lines. To this end, we found Secernin 1 (SCRN1), Lysyl oxidaselike 1 (LoxL1), Thymosin beta 4 X-linked (TMSB4X), Vesicleassociated membrane protein 5 (Vamp 5) and Serine protease inhibitor B1 (SerpinB1) as differential expressed in chemosensitive versus chemoresistant melanoma cells. Furthermore, we correlated the relative expression of our candidates with the chemosensitivity index of different chemotherapy regimes in up to 128 melanoma tissues so far. Importantly, we found a significant correlation between SerpinB1 gene but not protein expression and chemosensitivity towards Paclitaxel and Platin. Moreover, we also detected a differential expression of SerpinB1 in melanoma cell lines which however was running in reverse direction compared to the analyzed tissues. In conclusion, SerpinB1 seems to be a promising biomarker for prediction of Paclitaxel and Platin chemosensitivity. Merkel cell carcinoma (MCC) is a rare and highly aggressive skin cancer associated with the Merkel cell polyomavirus (MCV). As MCC cell lines demonstrate oncogene addiction to the MCV T-antigens, pharmacological interference of the large T-antigen(LTA) may represent an effective therapeutic approach for this deadly cancer. In this study, we investigated the effects of interferons (IFNs) on MCC cell lines, especially on MCV positive (MCV+) lines. Type I IFNs (i.e. Multiferon, a mix of different IFN α subtypes, and IFN β) strongly inhibited the cellular viability. Cell cycle analysis demonstrated increased sub-G fractions for these cells upon IFN treatment indicating apoptotic cell deathν these effects were less pronounced for IFN β. Notably, this inhibitory effect of type I IFNs on MCV+ MCC cell lines was associated with a reduced expression of the MCV LTA as well as an increased expression of promyelocytic leukemia protein (PML), which is known to interfere with the function of the LTA. In addition, the intra-tumoural application of multiferon resulted in a regression of MCV+ but not MCV- MCCs in vivo. Together, our findings demonstrate that type I IFNs have a strong antitumour effect, which is at least in part explained by modulation of the virally encoded LTA. Moreover, in addition to directly affecting MCC cell proliferation, IFNs strongly reinduce MHC class I expression in MCC cells. Flowcytometry demonstrated a re-induction of MHC class I expression upon IFN treatment in three MHC class I- MCV+ cell lines and an increase in MHC class I expression in cell lines that were characterized by a weak expression prior to treatment. Importantly, the increase or induction of MHC class I expression could also be demonstrated in vivo in xenotransplantation models. These results imply that IFN treatment has both a direct and an indirect effect in MCC and should be applicable in a general manner, i.e. irrespective of the MCV status of the patient. Beside IFN, Artemisinins are also known for their antitumoral and antiviral properties. In consequence, we tested the effect of Artesunate, i.e., an Artemisinin drivate, on MCV+ and MCV- MCC cell lines. In this regard, we could demonstrate an antiproliferative effect which was stronger on MCV+ cell lines, and which was associated with a reduced expression of the viral LTA. In contrast, fibroblasts were uneffected by Artenusate treatment. The reduced tumor growth could also be shown in vivo by intra-tumoral injection of Artesunate in MCV+ xenotransplantation models. According to these findings, a more detailed investigation of this ancient natural drug for the treatment of MCC patients should be considered. KW - Interferon KW - Melanom KW - Qinghaosu KW - Chemosensitivität KW - Merkelzellkarzinom KW - Chemosensitivity KW - Merkel cell carcinoma KW - Merkel-Zellkarzinom Y1 - 2013 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-83470 ER - TY - JOUR A1 - Sarma, Bhavishya A1 - Willmes, Christoph A1 - Angerer, Laura A1 - Adam, Christian A1 - Becker, Jürgen C. A1 - Kervarrec, Thibault A1 - Schrama, David A1 - Houben, Roland T1 - Artesunate affects T antigen expression and survival of virus-positive Merkel cell carcinoma JF - Cancers N2 - Merkel cell carcinoma (MCC) is a rare and highly aggressive skin cancer with frequent viral etiology. Indeed, in about 80% of cases, there is an association with Merkel cell polyomavirus (MCPyV); the expression of viral T antigens is crucial for growth of virus-positive tumor cells. Since artesunate — a drug used to treat malaria — has been reported to possess additional anti-tumor as well as anti-viral activity, we sought to evaluate pre-clinically the effect of artesunate on MCC. We found that artesunate repressed growth and survival of MCPyV-positive MCC cells in vitro. This effect was accompanied by reduced large T antigen (LT) expression. Notably, however, it was even more efficient than shRNA-mediated downregulation of LT expression. Interestingly, in one MCC cell line (WaGa), T antigen knockdown rendered cells less sensitive to artesunate, while for two other MCC cell lines, we could not substantiate such a relation. Mechanistically, artesunate predominantly induces ferroptosis in MCPyV-positive MCC cells since known ferroptosis-inhibitors like DFO, BAF-A1, Fer-1 and β-mercaptoethanol reduced artesunate-induced death. Finally, application of artesunate in xenotransplanted mice demonstrated that growth of established MCC tumors can be significantly suppressed in vivo. In conclusion, our results revealed a highly anti-proliferative effect of the approved and generally well-tolerated anti-malaria compound artesunate on MCPyV-positive MCC cells, suggesting its potential usage for MCC therapy. KW - artesunate KW - Merkel cell carcinoma KW - MCC KW - polyomavirus KW - ferroptosis Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-203851 SN - 2072-6694 VL - 12 IS - 4 ER - TY - THES A1 - Sarma, Bhavishya T1 - Merkel Cell Carcinoma: Investigations on its carcinogenesis and new therapeutic approaches T1 - Merkelzellkarzinom: Untersuchungen zur Karzinogenese und neue Therapieansätze N2 - Merkel cell carcinoma (MCC) is a rare and aggressive skin cancer with an increasing incidence. The majority of MCC cases (approximately 80%) are associated with the Merkel cell polyomavirus (MCPyV). This virus encodes for the MCPyV T antigens (small T (sT) and large T (LT)), which are oncoproteins that drive MCC carcinogenesis. However, the precise cells of the skin that are transformed by the T antigens are not known i.e., the cells of origin of MCC are yet to be discovered. Therefore, the first part of this study involved the generation and evaluation of a vector system that could be used to study MCC oncogenesis. To this end, a set of lentiviral vectors was cloned that allows independent, inducible expression of potential key factors in MCC oncogenesis. In addition, a CRISPR/Cas9 knock in was established that allows the coding sequence for a fluorescent protein to be placed under the control of the promoter of KRT20, one of the most crucial markers of MCC. The functionality of this KRT20 reporter was proven in the MCPyV-positive MCC cell line, WaGa. The different inducible vector systems (doxycycline-inducible MCPyV T antigens or MCPyV sT, RheoSwitch-inducible ATOH1 and IPTG-inducible dnMAML1 and GLI1) were found to have different efficacies in various cellular systems and in particular, a considerable reduction in efficiency was observed at times upon the interaction of several vectors in one cell. In the second and more important part of this study, the role of the well-established anti-malarial drug, artesunate, which possesses additional anti-tumor and anti-viral activity, in the treatment of MCPyV-positive MCC was analyzed. In our study, artesunate was found to be cytotoxic towards MCPyV-positive MCC cell lines in vitro and repressed tumor growth in vivo in a mouse model. Artesunate was also found to downregulate T antigen expression, which is critical for the proliferation of MCPyV-positive MCC cells. The repression of T antigen expression, however, was not the sole mechanism of artesunate’s cytotoxic action; instead, the MCPyV-positive MCC cell line, WaGa, was found to be even less sensitive to artesunate after shRNA knockdown of the T antigens. Since loss of membrane integrity occurred more rapidly than degradation/loss of genomic DNA under the influence of artesunate in four of five MCPyV-positive MCC cell lines examined, apoptosis, although widely described as a modus operandi for artesunate, did not appear to be a determinant of the cytotoxicity of artesunate against MCPyV-positive MCC cells. Instead, we were able to demonstrate that artesunate induced the recently described iron-dependent and lipid peroxide-associated form of cell death known as "ferroptosis". This was achieved primarily through the use of inhibitors that can suppress specific individual steps of the ferroptotic process. Thus, artesunate-induced cell death of MCPyV-positive MCC cells could be suppressed by iron chelators and by the inhibition of lipid peroxidation and lysosomal transport. Surprising results were obtained from the analysis of two proteins associated with the ferroptotic process, namely, ferroptosis suppressor protein 1 (FSP1) and tumor suppressor protein p53. Here, we showed that ectopically- 2 expressed FSP1 cannot suppress artesunate-induced ferroptosis in MCPyV-positive MCC cells and that p53 does not play a pro-ferroptotic role in artesunate-induced cell death of MCPyV-positive MCCs. Since artesunate did not suppress the interferon-γ-induced expression of immune-related molecules such as HLA and PD-L1 on the surface of MCPyV-positive MCCs, our study also provided the first positive evidence for its use in combinatorial immunotherapy. Overall, this study showed that artesunate appears to be an effective drug for the treatment of MCPyV-positive MCC and might also be considered for its use in combinatorial MCC immunotherapy in the future. N2 - Das Merkelzellkarzinom (MCC) ist ein seltener und aggressiver Hautkrebs mit einer steigenden Inzidenz. Die Mehrzahl der MCC-Fälle (ca. 80%) sind mit dem Merkelzell-Polyomavirus (MCPyV) assoziiert. Dieses Virus kodiert für die MCPyV T Antigene (small T (sT) und Large T (LT)), die als Onkoproteine die MCC Karzinogenese vorantreiben. Unbekannt ist jedoch welche Zellen der Haut durch die T Antigene transformiert werden, was also die Ursprungszellen des MCC sind. Der erste Teil dieser Arbeit befasste sich daher mit der Generierung und Evaluierung eines Vektorsystems, das zur Untersuchung der MCC-Onkogenese genutzt werden könnte. Dazu wurde ein Set von lentiviralen Vektoren kloniert, das die voneinander unabhängige, induzierbare Expression von potentiellen Schlüsselfaktoren der MCC-Onkogenese erlaubt. Außerdem wurde ein CRISPR/Cas9 knock in etabliert, der es erlaubt die kodierende Sequenz für ein fluoreszierendes Protein unter die Kontrolle des Promoters von KRT20, dessen Expression eines der wichtigsten MCC Marker ist, zu bringen. Die Funktionalität dieses KRT20 Reporters konnte in einer MCPyV-positive MCC Linie, WaGa, nachgewiesen werden. Die verschiedenen induzierbaren Vektorsysteme (Doxycyclin-induzierbare MCPyV T Antigene bzw. MCPyV sT, RheoSwitch-induzierbare ATOH1, IPTG-induzierbares dnMAML1 und GLI1) zeigten sich unterschiedlich effektiv in unterschiedlichen Zellsystemen und insbesondere im Zusammenspiel mehrerer Vektoren in einer Zelle war die Induzierbarkeit teilweise erheblich reduziert. Im zweiten, und wichtigeren Teil dieser Arbeit, habe ich mich mit der Frage auseinandergesetzt, ob ein etabliertes Anti-Malaria-Medikament, dem zusätzlich anti-tumorale und anti-virale Aktivität zugeschrieben werden, sich für die Behandlung des MCPyV-positiven MCC anbieten könnte. In unserer Studie wurde festgestellt, dass Artesunat in vitro zytotoxisch auf MCPyV-positive MCC Zelllinien wirkt und in vivo im Mausmodell das Tumorwachstum verlangsamen kann. Es wurde zudem beobachtet, dass Artesunat die T Antigen Expression unterdrückt, das für die Proliferation von MCPyV-positiven MCC-Zellen entscheidend ist. Allerdings spielt die Repression der T Antigen Expression keine Rolle für den zytotoxischen Effekt von Artesunat, sondern es zeigte sich, dass die MCC-Zelllinie WaGa nach shRNA knockdown der T Antigene sogar weniger sensitiv gegenüber Artesunat war. Da unter dem Einfluss von Artesunat in vier von fünf untersuchten MCC Zelllinien der Verlust der Membranintegrität schneller eintrat als eine Degradation/Verlust der genomischen DNA scheint Apoptose, obwohl vielfach als Modus Operandi für Artesunat beschrieben, nicht bestimmend für die Zytotoxität von Artesunat gegenüber MCPyV-positiven MCC Zellen zu sein. Stattdessen konnten wir die kürzlich beschriebene eisenabhängige und Lipidperoxid-assoziierte Form des Zelltods, die sogenannte "Ferroptose", nachweisen. Dies gelang vor allem durch den Einsatz von Inhibitoren, die spezifische Einzelschritte des ferroptotischen Prozesses unterdrücken können. So ließ sich der durch Artesunat induzierte Zelltod von MCPyV-positiven Zellen durch Eisenchelatbildner und Inhibition der Lipidperoxidation und des lysosomalen Transports unterdrücken. Überraschende Ergebnisse lieferte die Analyze von zwei Proteinen, die mit dem ferroptotischen Prozess in Verbindung gebracht werden, nämlich dem Ferroptose-Suppressor-Protein 1 (FSP1) und dem Tumorsuppressor-Protein p53. Hier zeigte sich, dass ektopisch-exprimiertes FSP1 die Artesunat-induzierte Ferroptose in MCC Zellen nicht unterdrücken kann und dass p53 keine pro-ferroptotische Rolle beim Artesunat-induzierten Zelltod von MCPyV-positiven MCCs spielt. Da Artesunat die Interferon-γ induzierte Expression von immunrelevanten Molekülen wie HLA und PD-L1 auf der Oberfläche von MCPyV-positiven MCCs nicht unterdrückte, ergaben sich auch erste positive Hinweise auf seinen Einsatz in der kombinatorischen Immuntherapie. Insgesamt zeigte diese Studie, dass Artesunat ein wirksames Medikament für die Behandlung von MCPyV-positivem MCC zu sein scheint und in Zukunft auch für den Einsatz in der kombinatorischen MCC-Immuntherapie in Frage kommen könnte. KW - Merkel cell carcinoma KW - Merkel cell polyomavirus KW - Ferroptosis KW - Artesunate Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-247402 ER - TY - JOUR A1 - Ritter, Cathrin A1 - Fan, Kaiji A1 - Paulson, Kelly G. A1 - Nghiem, Paul A1 - Schrama, David A1 - Becker, Jürgen C. T1 - Reversal of epigenetic silencing of MHC class I chain-related protein A and B improves immune recognition of Merkel cell carcinoma JF - Scientific Reports N2 - Merkel cell carcinoma (MCC) is a virally associated cancer characterized by its aggressive behavior and strong immunogenicity. Both viral infection and malignant transformation induce expression of MHC class I chain-related protein (MIC) A and B, which signal stress to cells of the immune system via Natural Killer group 2D (NKG2D) resulting in elimination of target cells. However, despite transformation and the continued presence of virally-encoded proteins, MICs are only expressed in a minority of MCC tumors in situ and are completely absent on MCC cell lines in vitro. This lack of MIC expression was due to epigenetic silencing via MIC promoter hypo-acetylation; indeed, MIC expression was re-induced by pharmacological inhibition of histone deacetylases (HDACs) both in vitro and in vivo. This re-induction of MICs rendered MCC cells more sensitive to immune-mediated lysis. Thus, epigenetic silencing of MICs is an important immune escape mechanism of MCCs. KW - epigenetic silencing KW - Merkel cell carcinoma KW - MHC class I chain-related protein KW - skin cancer Y1 - 2016 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-167992 IS - 21678 ET - 6 ER - TY - THES A1 - Ritter, Cathrin T1 - Scientific basics for new immunotherapeutic approaches towards Merkel cell carcinoma T1 - Grundlagen neuer immuntherapeutischer Ansätze gegen das Merkelzellkarzinom N2 - Merkel cell carcinoma (MCC) is an aggressive neuroendocrine skin cancer that has been associated with the Merkel cell polyomavirus (MCPyV). Indeed, MCC is one of the cancers with the best-established viral carcinogenesis. Despite persistence of the virus in MCC cells and the subsequent expression of viral antigens, the majority of MCC tumors are able to escape the surveillance of the immune system. Therefore the aim of the here presented thesis was to scrutinize immune escape mechanisms operative in MCC. A better understanding of their underlying molecular processes should allow to improve immunotherapeutic treatment strategies for MCC patients. The manuscripts included in this thesis characterize three novel immune evasion strategies of MCC. I) the epigenetic silencing of the NKG2D ligands MICA and MICB via histone H3 hypoacetylation II) reduced HLA class I surface expression via epigenetic silencing of the antigen processing machinery (APM) III) the activation of the PI3K-AKT pathway in a mutation independent manner as potential immune escape strategy MCC tumors and MCC cell lines were analyzed for their expression of MICA/B, HLA and components of the antigen processing machinery as well as for the activation of the PI3K-AKT pathway in situ and in vitro. These analysis reviled MICA and MICB, as well as HLA class I were not expressed or at least markedly reduced in ~80% of MCCs in situ. The PI3K-AKT pathway, that had only recently been demonstrated to play a significant role in tumor immune escape, was activated in almost 90% of MCCs in situ. To determine the underlying molecular mechanisms of these aberrations well characterized MCC cell lines were further analyzed in vitro. The fact that the PI3K-AKT pathway activation was due to oncogenic mutations in the PIK3CA or AKT1 gene in only 10% of MCCs, suggested an epigenetic regulation of this pathway in MCC. In line with this MICA/B as well as components of the APM were indeed silenced epigenetically via histone hypoacetylation in their respective promoter region. Notably MICA/B and HLA class I expression on the cell surface of MCC cells could be restored after treatment with HDAC inhibitors in combination with the Sp1 inhibitor Mithramycin A in all analyzed MCC cell lines in vitro and in a xenotransplantation mouse model in vivo. Moreover inhibition of HDACs increased immune recognition of MCC cell lines in a MICA/B and HLA class I dependent manner. Several studies have accumulated evidence that immunotherapy is a promising treatment option for MCC patients due to the exquisite immunogenicity of this malignancy. However, current immunotherapeutic interventions towards solid tumors like MCC have to account for the plentitude of tumor immune escape strategies, in order to increase response rates. The immune escape mechanisms of MCC described in this thesis can be reverted by HDAC inhibition, thus providing the rationale to combine ‘epigenetic priming’ with currently tested immunotherapeutic regimens. N2 - Das Merkelzellkarzinom (MCC) ist ein aggressiver neuroendokriner Hautkrebs, der mit dem Merkelzell-Polyomavirus (MCPyV) assoziiert ist. Das MCC ist eine der Krebserkrankungen mit der am besten etablierten viralen Karzinogenese. Trotz der Anwesenheit des MCPyV in MCC-Zellen und der daraus einhergehenden Expression viraler Antigene sind die meisten MCC-Tumoren in der Lage der Überwachung durch das Immunsystem zu entgehen. Aus diesem Grund war das Ziel der hier vorliegenden Arbeit, neue im MCC operative „immune escape“ Mechanismen zu ermitteln. Ein besseres Verständnis der hierbei zugrunde liegenden Mechanismen, sollte es ermöglichen, immuntherapeutische Behandlungsstrategien für MCC-Patienten zu verbessern. Die vorgestellten Manuskripte beschreiben drei neuartige „immune evasion“ Strategien des MCC: I) die epigenetische Inaktivierung der NKG2D-Liganden MICA und MICB mittels Histone-H3-Hypoacetylierung II) eine reduzierte HLA Klasse I-Oberflächenexpression aufgrund epigenetischer Inaktivierung der Antigenprozessierungsmaschinerie (APM) III) die mutationsunabhängige Aktivierung des PI3K-AKT-Signalweges, als potentieller „immune escape“ Mechanismus MCC-Tumoren und MCC-Zelllinien wurden sowohl bezüglich der Expression von MICA/B, HLA Klasse I und Komponenten der APM als auch auf die Aktivierung des PI3K Signalweges in situ und in vitro untersucht. Diese Analysen zeigten, dass sowohl MICA und MICB als auch HLA Klasse I in ca. 80% der MCC-Tumoren in situ nicht, oder nur sehr reduziert, exprimiert wurden. Der PI3K-AKT-Signalweg, welcher erst kürzlich mit Tumor „immune escape“ in Verbindung gebracht wurde, war in fast 90% aller MCC-Tumoren in situ aktiviert. Um die zugrunde liegenden molekularen Mechanismen dieser Aberrationen zu entschlüsseln, wurden gut charakterisierte MCC-Zelllinien in vitro untersucht. Die Tatsache, dass der PI3K-AKT-Signalweg in nur 10% der MCCs auf Mutationen im PI3KA- oder AKT1-Gen zurückzuführen war, suggeriert eine epigenetische Regulation dieses Signalwegs. In Übereinstimmung hiermit waren sowohl MICA/B als auch Gene der APM epigenetisch mittels Histon-Hypoacetylierung in ihren jeweiligen Promoterregionen inaktiviert. Bemerkenswerterweise konnten in vitro und in einem Xenotransplantations-Mausmodell in vivo sowohl die MICA/B als auch die HLA Klasse I-Oberflächenexpression aller untersuchter MCC-Zelllinien durch die Behandlung mit HDAC-Inhibitoren in Kombination mit dem Sp1-Inhibitor Mithramycin A wieder hergestellt werden. Des Weiteren erhöhte die Inhibition von HDACs die MCC-Immunerkennung auf eine MICA/B und HLA Klasse I-abhängige Weise. Zahlreiche aktuelle Studien bestärken die Annahme, dass aufgrund der besonderen Immunogenität des MCC, die Immuntherapie eine aussichtsreiche Behandlungsoption für MCC-Patienten darstellt. Nichtsdestotrotz müssen die derzeitigen immuntherapeutischen Methoden zur Behandlung solider Tumore die Vielzahl von Tumor „immune escape“ Mechanismen mitberücksichtigen, um die Ansprechrate zu erhöhen. Die Tatsache, dass die hier beschriebenen „immune escape“ Mechanismen durch HDAC-Inhibition aufgehoben werden können, spricht für die Hypothese, dass eine Kombination von „epigenetischem Priming“ mit derzeitig untersuchten immuntherapeutischen Ansätzen sinnvoll ist. KW - Merkel-Zellkarzinom KW - Merkel cell carcinoma KW - Epigenetics KW - MICA KW - MICB KW - Tumor Immunology KW - Immune Escape Y1 - 2017 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-124162 ER - TY - JOUR A1 - Orouji, Elias A1 - Peitsch, Wiebke K. A1 - Orouji, Azadeh A1 - Houben, Roland A1 - Utikal, Jochen T1 - Unique role of histone methyltransferase PRDM8 in the tumorigenesis of virus-negative Merkel cell carcinoma JF - Cancers N2 - Merkel cell carcinoma (MCC) is a deadly skin cancer, and about 80% of its cases have been shown to harbor integrated Merkel polyomavirus in the tumor cell genome. Viral oncoproteins expressed in the tumor cells are considered as the oncogenic factors of these virus-positive Merkel cell carcinoma (VP-MCC). In contrast, the molecular pathogenesis of virus-negative MCC (VN-MCC) is less well understood. Using gene expression analysis of MCC cell lines, we found histone methyltransferase PRDM8 to be elevated in VN-MCC. This finding was confirmed by immunohistochemical analysis of MCC tumors, revealing that increased PRDM8 expression in VN-MCC is also associated with increased H3K9 methylation. CRISPR-mediated silencing of PRDM8 in MCC cells further supported the histone methylating role of this protein in VN-MCC. We also identified miR-20a-5p as a negative regulator of PRDM8. Taken together, our findings provide insights into the role of PRDM8 as a histone methyltransferase in VN-MCC tumorigenesis. KW - histone KW - histone methyltransferase PRDM8 KW - chromatin regulator KW - EGR1 KW - miRNA KW - MCV-negative KW - Merkel cell carcinoma KW - MCPyV Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-203815 SN - 2072-6694 VL - 12 IS - 4 ER - TY - JOUR A1 - Orouji, Elias A1 - Peitsch, Wiebke K. A1 - Orouji, Azadeh A1 - Houben, Roland A1 - Utikal, Jochen T1 - Oncogenic role of an epigenetic reader of m\(^6\)A RNA modification: YTHDF1 in Merkel cell carcinoma JF - Cancers N2 - Merkel cell carcinoma is a deadly skin cancer, which in the majority of cases is caused by the Merkel cell polyomavirus (MCPyV). The viral small T antigen is regarded as the dominant oncoprotein expressed in the tumor cells. We used genomic screening of copy number aberrations along with transcriptomic analysis to investigate regions with amplification that harbor differentially expressed genes. We identified YTHDF1, a protein that is a reader of N\(^6\)-methyladenosine (m\(^6\)A) RNA modifications, to have high copy gains and to be highly expressed in Merkel cell carcinoma. Importantly, we identified the presence of m\(^6\)A on small T antigen mRNA suggesting a relation between YTHDF1 amplification and MCPyV gene expression. Interestingly, knockdown of YTHDF1 in Merkel cell carcinoma (MCC) cell lines negatively affected the translation initiation factor eIF3 and reduced proliferation and clonogenic capacity in vitro. Furthermore, analysis of survival data revealed worse overall survival in YTHDF1\(^{high}\) MCC patients compared to YTHDF1\(^{low}\) patients. Our findings indicate a novel oncogenic role of YTHDF1 through m\(^6\)A machinery in the tumorigenesis of MCC. KW - Merkel cell carcinoma KW - Merkel cell polyomavirus KW - copy number variations KW - m\(^6\)A KW - RNA modification KW - epitranscriptome KW - YTHDF1 KW - epigenetic reader Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-200815 SN - 2072-6694 VL - 12 IS - 1 ER - TY - THES A1 - Kleffel, Sonja Beate T1 - The role of cancer cell-expressed PD-1 in tumorigenesis and tumor immune evasion T1 - Funktionelle Charakterisierung von Tumorzell-exprimiertem PD-1 in der Karzinogenese und antitumoralen Immunabwehr N2 - Melanoma and Merkel cell carcinoma (MCC) are highly aggressive cancers of the skin that frequently escape immune recognition and acquire resistance to chemotherapeutic agents, which poses a major obstacle to successful cancer treatment. Recently, a new class of therapeutics targeting the programmed cell death-1 (PD-1) immune checkpoint receptor has shown remarkable efficacy in the treatment of both cancers. Blockade of PD-1 on T cells activates cancer-specific immune responses that can mediate tumor regression. The data presented in this Ph.D. thesis demonstrates that PD-1 is also expressed by subsets of cancer cells in melanoma and MCC. Moreover, this work identifies PD-1 as a novel tumor cell-intrinsic growth receptor, even in the absence of T cell immunity. PD-1 is expressed by tumorigenic cell subsets in melanoma patient samples and established human and murine cell lines that also co-express ABCB5, a marker of immunoregulatory tumor- initiating cells in melanoma. Consistently, melanoma-expressed PD-1 downmodulates T effector cell functions and increases the intratumoral frequency of tolerogenic myeloid- derived suppressor cells. PD-1 inhibition on melanoma cells by RNA interference, blocking antibodies, or mutagenesis of melanoma-PD-1 signaling motifs suppresses tumor growth in immunocompetent, immunocompromised, and PD-1-deficient tumor graft recipient mice. Conversely, melanoma-specific PD-1 overexpression enhances tumorigenicity, including in mice lacking adaptive immunity. Engagement of melanoma- PD-1 by its ligand PD-L1 promotes tumor growth, whereas melanoma-PD-L1 inhibition or knockout of host-PD-L1 attenuates growth of PD-1-positive melanomas. Mechanistically, the melanoma-PD-1 receptor activates mTOR signaling mediators, including ribosomal protein S6. In a proof-of-concept study, tumoral expression of phospho-S6 in pretreatment tumor biopsies correlated with clinical responses to anti-PD-1 therapy in melanoma patients. In MCC, PD-1 is similarly co-expressed by ABCB5+ cancer cell subsets in clinical tumor specimens and established human cell lines. ABCB5 renders MCC cells resistant to the standard-of-care chemotherapeutic agents, carboplatin and etoposide. Antibody-mediated ABCB5 blockade reverses chemotherapy resistance and inhibits tumor xenograft growth by enhancing chemotherapy-induced tumor cell killing. Furthermore, engagement of MCC-expressed PD-1 by its ligands, PD-L1 and PD-L2, promotes proliferation and activates MCC-intrinsic mTOR signaling. Consistently, antibody- mediated PD-1 blockade inhibits MCC tumor xenograft growth and phosphorylation of mTOR effectors in immunocompromised mice. In summary, these findings identify cancer cell-intrinsic functions of the PD-1 pathway in tumorigenesis and suggest that blocking melanoma- and MCC-expressed PD-1 might contribute to the striking clinical efficacy of anti-PD-1 therapy. Additionally, these results establish ABCB5 as a previously unrecognized chemoresistance mechanism in MCC. N2 - Das Melanom und das Merkelzellkarzinom (MZK) sind auttumoren neuroendokrinen Ursprungs, die sich durch ein besonders aggressives Wachstum auszeichnen. Melanome und MZK entgehen häufig der antitumoralen Immunabwehr und erwerben Resistenzen gegen Chemotherapeutika, was eine erfolgreiche Behandlung der betroffenen Patienten erschwert. In klinischen Studien hat eine neue Klasse von therapeutischen Antikörpern, die den Immun-Checkpoint Rezeptor PD-1 (Programmed Cell Death-1) inhibieren, hohe Ansprechraten und dauerhafte Remissionen bei Melanom- und MZK-Patienten erzielt. Die Blockade des PD-1 Rezeptors auf T-Zellen reaktiviert autologe Immunreaktionen gegen Tumorzellen, die zur Reduktion des Tumors führen können. Die vorgelegte Dissertation zeigt, dass Subpopulationen von Melanom- und MZK-Zellen PD-1 exprimieren, und dass die Aktivierung von Tumorzell-intrinsischem PD-1 einen pro-tumorigenen Mechanismus darstellt, einschliesslich in T-Zell-defizienten Mäusen. In Biopsien von Melanom-Patienten, sowie in humanen und murinen Melanom-Zelllinien wird PD-1 präferentiell von tumorigenen, immunregulatorischen, ABCB5+ Melanom-Stammzellen exprimiert. PD-1+ Melanomzellen hemmen die Aktivität von Effektor-T-Zellen und erhöhen die Anzahl der tolerogenen myeloiden Suppressorzellen im Tumor. Die Inhibierung des PD-1 Rezeptors auf Melanomzellen durch RNA-Interferenz, blockierende Antikörper oder Mutagenese der intrazellulären Signalmotive des PD-1 Proteins unterdrückt das Melanom-Wachstum in immunkompetenten, immunsupprimierten und PD-1-defizienten Mäusen. Umgekehrt führt die Melanom-spezifische Überexpression von PD-1 zu einem signifikant erhöhtem Tumorwachstum, sogar in immunsupprimierten Mäusen. Die Aktivierung des PD-1 Rezeptors auf Melanomzellen durch die Bindung seines Liganden, PD-L1, fördert das Tumorwachstum, während das protumorigene Potential von PD-1-positiven Melanomzellen durch die Inhibierung von PD-L1 auf Melanomzellen, sowie in PD-L1-defizienten Mäusen, gehemmt wird. In Melanomzellen aktiviert der PD-1 Rezeptor den mTOR Signaltransduktionsweg, einschließlich des Effektormoleküls ribosomales Protein S6. In einer Teststudie korrelierte die Expression des Phospho-S6 Proteins in Melanomzellen aus Biopsien, die vor Gabe der Immuntherapie entnommen wurden, mit den Ansprechraten der Melanom Patienten auf die Behandlung mit PD-1-Antikörpern. Auch in Biopsien von MZK-Patienten und in etablierten humanen MZK-Zelllinien wird PD-1 präferentiell von ABCB5+ Subpopulationen exprimiert. Im MZK vermittelt der ABCB5-Membrantransporter Resistenzen gegenüber den Zytostatika Carboplatin und Etoposid. Die Antikörper-vermittelte Blockade des ABCB5-Transporters sensibilisiert MZK-Zellen für die Carboplatin- und Etoposid-vermittelte Apoptose, was zu einer signifikanten Reduktion des experimentellen Tumorwachstums führt. Ähnlich wie im Melanom fördert die Bindung des PD-1 Rezeptors auf MZK Zellen durch seine Liganden, PD-L1 und PD-L2, deren Proliferation und die intrazelluläre Aktivierung der mTORSignalkaskade. Entsprechend führt die antikörper-vermittelte Blockade von PD-1 zur Inhibierung des MZK-Tumorwachstums in immunsupprimierten Mäusen und zu einer reduzierten Phosphorylierung von mTOR Effektormolekülen. Zusammenfassend konnte in der vorliegenden Dissertation gezeigt werden, dass Subpopulationen von Melanom- und MZK-Zellen PD-1 exprimieren, und dass Tumorzell-intrinsische PD-1-Funktionen das Krebswachstum fördern. Diese Ergebnisse deuten darauf hin, dass die Blockade des PD-1-Rezeptors auf Tumorzellen zu der klinischen Wirksamkeit der anti-PD-1 Therapie beitragen könnte. Darüber hinaus konnte ABCB5 als neuer Chemoresistenz-Mechanismus in MZK identifiziert werden. KW - Melanom KW - Merkelzellkarzinom KW - Cancer KW - Melanoma KW - Merkel cell carcinoma KW - Cancer immunotherapy KW - Chemotherapeutic resistance Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-151205 ER - TY - THES A1 - Kervarrec, Thibault T1 - Histogenesis of Merkel cell carcinoma T1 - Histogenese des Merkelzellkarzinoms N2 - Merkel cell carcinoma (MCC) is a rare and aggressive skin cancer. In approximately 80% of cases, genomic integration of the Merkel cell polyomavirus (MCPyV) is observed and overexpression of the two MCPyV T antigens (TAgs) is regarded as the main oncogenic determinant of MCPyV-positive MCC cases. However, the nature of the cells from which MCC arises is unknown. Therefore, the goal of the present work was to determine the cell of origin of MCC. First, we characterized MCC patients’ tumors and demonstrated a high similarity of MCPyV- negative MCC with extracutaneous neuroendocrine carcinoma while MCPyV-positive MCC differs from these two groups with respect to morphology, immunohistochemical profile, genetics, origin and behavior. Based on the analysis of a trichoblastoma/MCC combined tumor, we demonstrated that a MCPyV-positive MCC can arise following MCPyV integration in an epithelial cell. In addition, the high similarity between trichoblastoma cells and Merkel cell (MC) progenitors of the hair follicle suggests that these hair follicle cells may represent a general start point for the development of MCPyV-positive MCC. A contribution of the viral TAgs to the development of the characteristic Merkel cell-like MCC phenotype is suggested by experiments demonstrating induction of Merkel cell markers upon TAg expression in human primary keratinocytes or hair follicle cells. As potential mechanisms mediating these phenotypic changes, we identified the capability of MCPyV LT to repress degradation of master regulator of MC development, i.e. the transcription factor ATOH1. To conclude, our work suggests that MCPyV integration in epithelial cells of the hair follicle may represent an important path for MCC development. N2 - Das Merkelzellkarzinom (MCC) ist ein seltener und aggressiver Hautkrebs. In etwa 80% der Fälle wird die genomische Integration des Merkelzell-Polyomavirus (MCPyV) beobachtet und die Überexpression der beiden MCPyV-T-Antigene (TAgs) gilt als die wichtigste onkogene Determinante der MCPyV-positiven MCC-Fälle. Die Ursprungszelle des MCC ist jedoch bisher unbekannt. Daher war das Ziel der vorliegenden Arbeit, die Hinweise auf die Herkunftszelle zu generieren. ... KW - Merkel-Zellkarzinom KW - Merkel cell carcinoma KW - Histogenesis KW - Hair follicle KW - Polyomavirus KW - Histogenese Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-199750 ER - TY - JOUR A1 - Houben, Roland A1 - Ebert, Marlies A1 - Hesbacher, Sonja A1 - Kervarrec, Thibault A1 - Schrama, David T1 - Merkel Cell Polyomavirus Large T Antigen is Dispensable in G2 and M-Phase to Promote Proliferation of Merkel Cell Carcinoma Cells JF - Viruses N2 - Merkel cell carcinoma (MCC) is an aggressive skin cancer frequently caused by the Merkel cell polyomavirus (MCPyV), and proliferation of MCPyV-positive MCC tumor cells depends on the expression of a virus-encoded truncated Large T antigen (LT) oncoprotein. Here, we asked in which phases of the cell cycle LT activity is required for MCC cell proliferation. Hence, we generated fusion-proteins of MCPyV-LT and parts of geminin (GMMN) or chromatin licensing and DNA replication factor1 (CDT1). This allowed us to ectopically express an LT, which is degraded either in the G1 or G2 phase of the cell cycle, respectively, in MCC cells with inducible T antigen knockdown. We demonstrate that LT expressed only in G1 is capable of rescuing LT knockdown-induced growth suppression while LT expressed in S and G2/M phases fails to support proliferation of MCC cells. These results suggest that the crucial function of LT, which has been demonstrated to be inactivation of the cellular Retinoblastoma protein 1 (RB1) is only required to initiate S phase entry. KW - Merkel cell polyomavirus KW - large T antigen KW - cell cycle KW - Merkel cell carcinoma Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-218171 SN - 1999-4915 VL - 12 IS - 10 ER - TY - JOUR A1 - Houben, Roland A1 - Celikdemir, Büke A1 - Kervarrec, Thibault A1 - Schrama, David T1 - Merkel cell polyomavirus: infection, genome, transcripts and its role in development of Merkel cell carcinoma JF - Cancers N2 - The best characterized polyomavirus family member, i.e., simian virus 40 (SV40), can cause different tumors in hamsters and can transform murine and human cells in vitro. Hence, the SV40 contamination of millions of polio vaccine doses administered from 1955–1963 raised fears that this may cause increased tumor incidence in the vaccinated population. This is, however, not the case. Indeed, up to now, the only polyomavirus family member known to be the most important cause of a specific human tumor entity is Merkel cell polyomavirus (MCPyV) in Merkel cell carcinoma (MCC). MCC is a highly deadly form of skin cancer for which the cellular origin is still uncertain, and which appears as two clinically very similar but molecularly highly different variants. While approximately 80% of cases are found to be associated with MCPyV the remaining MCCs carry a high mutational load. Here, we present an overview of the multitude of molecular functions described for the MCPyV encoded oncoproteins and non-coding RNAs, present the available MCC mouse models and discuss the increasing evidence that both, virus-negative and -positive MCC constitute epithelial tumors. KW - Merkel cell carcinoma KW - polyomavirus KW - T antigen Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-305021 SN - 2072-6694 VL - 15 IS - 2 ER - TY - JOUR A1 - Houben, Roland A1 - Alimova, Pamela A1 - Sarma, Bhavishya A1 - Hesbacher, Sonja A1 - Schulte, Carolin A1 - Sarosi, Eva-Maria A1 - Adam, Christian A1 - Kervarrec, Thibault A1 - Schrama, David T1 - 4-[(5-methyl-1H-pyrazol-3-yl)amino]-2H-phenyl-1-phthalazinone inhibits MCPyV T antigen expression in Merkel cell carcinoma independent of Aurora kinase A JF - Cancers N2 - Merkel cell carcinoma (MCC) is frequently caused by the Merkel cell polyomavirus (MCPyV), and MCPyV-positive tumor cells depend on expression of the virus-encoded T antigens (TA). Here, we identify 4-[(5-methyl-1H-pyrazol-3-yl)amino]-2H-phenyl-1-phthalazinone (PHT) — a reported inhibitor of Aurora kinase A — as a compound inhibiting growth of MCC cells by repressing noncoding control region (NCCR)-controlled TA transcription. Surprisingly, we find that TA repression is not caused by inhibition of Aurora kinase A. However, we demonstrate that β-catenin — a transcription factor repressed by active glycogen synthase kinase 3 (GSK3) — is activated by PHT, suggesting that PHT bears a hitherto unreported inhibitory activity against GSK3, a kinase known to function in promoting TA transcription. Indeed, applying an in vitro kinase assay, we demonstrate that PHT directly targets GSK3. Finally, we demonstrate that PHT exhibits in vivo antitumor activity in an MCC xenograft mouse model, suggesting a potential use in future therapeutic settings for MCC. KW - Merkel cell carcinoma KW - polyomavirus KW - large T antigen KW - phthalazinone pyrazole KW - glycogen synthase kinase 3 KW - GSK3 Y1 - 2023 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-313547 SN - 2072-6694 VL - 15 IS - 9 ER - TY - JOUR A1 - Hesbacher, Sonja A1 - Pfitzer, Lisa A1 - Wiedorfer, Katharina A1 - Angermeyer, Sabrina A1 - Borst, Andreas A1 - Haferkamp, Sebastian A1 - Scholz, Claus-Jürgen A1 - Wobser, Marion A1 - Schrama, David A1 - Houben, Roland T1 - RB1 is the crucial target of the Merkel cell polyomavirus Large T antigen in Merkel cell carcinoma cells JF - Oncotarget N2 - The pocket protein (PP) family consists of the three members RB1, p107 and p130 all possessing tumor suppressive properties. Indeed, the PPs jointly control the G1/S transition mainly by inhibiting E2F transcription factors. Notably, several viral oncoproteins are capable of binding and inhibiting PPs. Merkel cell polyomavirus (MCPyV) is considered as etiological factor for Merkel cell carcinoma (MCC) with expression of the viral Large T antigen (LT) harboring an intact PP binding domain being required for proliferation of most MCC cells. Therefore, we analyzed the interaction of MCPyV-LT with the PPs. Co-IP experiments indicate that MCPyV-LT binds potently only to RB1. Moreover, MCPyV-LT knockdown-induced growth arrest in MCC cells can be rescued by knockdown of RB1, but not by p107 or p130 knockdown. Accordingly, cell cycle arrest and E2F target gene repression mediated by the single PPs can only in the case of RB1 be significantly reverted by MCPyV-LT expression. Moreover, data from an MCC patient indicate that loss of RB1 rendered the MCPyV-positive MCC cells LT independent. Thus, our results suggest that RB1 is the dominant tumor suppressor PP in MCC, and that inactivation of RB1 by MCPyV-LT is largely sufficient for its growth supporting function in established MCPyV-positive MCC cells. KW - Merkel cell carcinoma KW - polyomavirus KW - Large T antigen KW - retinoblastoma protein KW - viral carcinogenesis Y1 - 2016 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-177858 VL - 7 IS - 22 ER - TY - JOUR A1 - Glutsch, Valerie A1 - Kneitz, Hermann A1 - Gesierich, Anja A1 - Goebeler, Matthias A1 - Haferkamp, Sebastian A1 - Becker, Jürgen C. A1 - Ugurel, Selma A1 - Schilling, Bastian T1 - Activity of ipilimumab plus nivolumab in avelumab-refractory Merkel cell carcinoma JF - Cancer Immunology, Immunotherapy N2 - Background Merkel cell carcinoma (MCC) is a rare and aggressive neuroendocrine cutaneous malignancy with poor prognosis. In Europe, approved systemic therapies are limited to the PD-L1 inhibitor avelumab. For avelumab-refractory patients, efficient and safe treatment options are lacking. Methods At three different sites in Germany, clinical and molecular data of patients with metastatic MCC being refractory to the PD-L1 inhibitor avelumab and who were later on treated with combined IPI/NIVO were retrospectively collected and evaluated. Results Five patients treated at three different academic sites in Germany were enrolled. Three out of five patients investigated for this report responded to combined IPI/NIVO according to RECIST 1.1. Combined immunotherapy was well tolerated without any grade II or III immune-related adverse events. Two out of three responders to IPI/NIVO received platinum-based chemotherapy in between avelumab and combined immunotherapy. Conclusion In this small retrospective study, we observed a high response rate and durable responses to subsequent combined immunotherapy with IPI/NIVO in avelumab-refractory metastatic MCC patients. In conclusion, our data suggest a promising activity of second- or third-line PD-1- plus CTLA-4-blockade in patients with anti-PD-L1-refractory MCC. KW - ipilimumab KW - Merkel cell carcinoma KW - resistance KW - avelumab KW - nivolumab Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-265635 SN - 14320851 VL - 70 IS - 7 ER - TY - JOUR A1 - Fan, Kaiji A1 - Zebisch, Armin A1 - Horny, Kai A1 - Schrama, David A1 - Becker, Jürgen C. T1 - Highly expressed miR-375 is not an intracellular oncogene in Merkel cell polyomavirus-associated Merkel cell carcinoma JF - Cancers N2 - miR-375 is a highly abundant miRNA in Merkel cell carcinoma (MCC). In other cancers, it acts as either a tumor suppressor or oncogene. While free-circulating miR-375 serves as a surrogate marker for tumor burden in patients with advanced MCC, its function within MCC cells has not been established. Nearly complete miR-375 knockdown in MCC cell lines was achieved using antagomiRs via nucleofection. The cell viability, growth characteristics, and morphology were not altered by this knockdown. miR-375 target genes and related signaling pathways were determined using Encyclopedia of RNA Interactomes (ENCORI) revealing Hippo signaling and epithelial to mesenchymal transition (EMT)-related genes likely to be regulated. Therefore, their expression was analyzed by multiplexed qRT-PCR after miR-375 knockdown, demonstrating only a limited change in expression. In summary, highly effective miR-375 knockdown in classical MCC cell lines did not significantly change the cell viability, morphology, or oncogenic signaling pathways. These observations render miR-375 an unlikely intracellular oncogene in MCC cells, thus suggesting that likely functions of miR-375 for the intercellular communication of MCC should be addressed. KW - miR-375 KW - antagomiRs KW - Merkel cell carcinoma KW - Hippo signaling KW - focal adhesion Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-200678 SN - 2072-6694 VL - 12 IS - 3 ER - TY - JOUR A1 - Buder, Kristina A1 - Lapa, Constantin A1 - Kreissl, Michael C. A1 - Schirbel, Andreas A1 - Herrmann, Ken A1 - Schnack, Alexander A1 - Bröcker, Eva-Bettina A1 - Goebeler, Matthias A1 - Buck, Andreas K. A1 - Becker, Jürgen C. T1 - "Somatostatin receptor expression in Merkel cell carcinoma as target for molecular imaging" N2 - Background Merkel cell carcinoma (MCC) is a rare cutaneous neoplasm with increasing incidence, aggressive behavior and poor prognosis. Somatostatin receptors (SSTR) are expressed in MCC and represent a potential target for both imaging and treatment. Methods To non-invasively assess SSTR expression in MCC using PET and the radiotracers [68Ga]DOTA-D-Phe1-Tyr3-octreotide (DOTATOC) or -octreotate (DOTATATE) as surrogate for tumor burden. In 24 patients with histologically proven MCC SSTR-PET was performed and compared to results of computed tomography (CT). Results SSTR-PET detected primary and metastatic MCC lesions. On a patient-based analysis, sensitivity of SSTR-PET was 73% for nodal metastases, 100% for bone, and 67% for soft-tissue metastases, respectively. Notably, brain metastases were initially detected by SSTR-PET in 2 patients, whereas liver and lung metastases were diagnosed exclusively by CT. SSTR-PET showed concordance to CT results in 20 out of 24 patients. Four patients (17%) were up-staged due to SSTR-PET and patient management was changed in 3 patients (13%). Conclusion SSTR-PET showed high sensitivity for imaging bone, soft tissue and brain metastases, and particularly in combination with CT had a significant impact on clinical stage and patient management. KW - Merkel cell carcinoma KW - Molecular imaging KW - Somatostatin receptor expression KW - Positron emission tomography Y1 - 2014 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-110326 ER - TY - JOUR A1 - Behr, Daniel S. A1 - Peitsch, Wiebke K. A1 - Hametner, Christian A1 - Lasitschka, Felix A1 - Houben, Roland A1 - Schönhaar, Kathrin A1 - Michel, Julia A1 - Dollt, Claudia A1 - Goebeler, Matthias A1 - Marx, Alexander A1 - Goerdt, Sergij A1 - Schmieder, Astrid T1 - Prognostic value of immune cell infiltration, tertiary lymphoid structures and PD-L1 expression in Merkel cell carcinomas JF - International Journal of Clinical and Experimental Pathology N2 - Merkel cell carcinoma (MCC) is an aggressive, virus-associated, neuroendocrine tumor of the skin mainly affecting immunocompromised patients. Higher intratumoral infiltration with CD3 and CD8 positive T-cells is associated with a better prognosis, highlighting the relevance of the immune system for MCC development and progression. In this study 21 primary MCCs were stained with immune cell markers including CD3, CD4, CD8, CD68, CD20, and S100. Furthermore, tumor-infiltrating neutrophils, tertiary lymphoid structures and PD-L1 expression were analyzed and correlated with overall and recurrence free survival. All MCCs were Merkel Cell Polyomavirus positive. Overall and recurrence-free survival did not correlate with intra-and peritumoral CD3 and CD8 T-cell infiltration. In addition, no significant association regarding prognosis was found for tumor-associated neutrophils, tumor-associated macrophages or PD-L1 positivity in MCCs. Interestingly, the presence of tertiary lymphoid structures (TLS) in the tumor microenvironment significantly correlated with recurrence-free survival (P=0.025). In addition, TLS were significantly associated with a higher CD8/CD4 ratio in the tumor periphery (P=0.032), but not in the center of the tumor (P > 0.999). These results demonstrate for the first time that TLS, easily assessed in paraffin-embedded tissue in the tumor periphery of MCCs, may be a valuable prognostic factor indicating prolonged recurrence free survival. KW - CD8(+) KW - PD-L1 KW - tertiary lymphoid structures KW - immune cell infiltration KW - polymavirus KW - survival KW - lymphocytes KW - responses KW - lung cancer KW - B-cells KW - breast cancer KW - antitumor immunity KW - T-antigens KW - Merkel cell carcinoma Y1 - 2014 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-117720 SN - 1936-2625 VL - 7 IS - 11 ER -