Dokument-ID Dokumenttyp Verfasser/Autoren Herausgeber Haupttitel Abstract Auflage Verlagsort Verlag Erscheinungsjahr Seitenzahl Schriftenreihe Titel Schriftenreihe Bandzahl ISBN Quelle der Hochschulschrift Konferenzname Quelle:Titel Quelle:Jahrgang Quelle:Heftnummer Quelle:Erste Seite Quelle:Letzte Seite URN DOI Abteilungen OPUS4-12509 Wissenschaftlicher Artikel Alb, Miriam; Sie, Christopher; Adam, Christian; Chen, Suzie; Becker, Jürgen C.; Schrama, David Cellular and cytokine-dependent immunosuppressive mechanisms of grm1-transgenic murine melanoma Grm1-transgenic mice spontaneously develop cutaneous melanoma. This model allowed us to scrutinize the generic immune responses over the course of melanoma development. To this end, lymphocytes obtained from spleens, unrelated lymph nodes and tumor-draining lymph nodes of mice with no evidence of disease, and low or high tumor burden were analyzed ex vivo and in vitro. Thereby, we could demonstrate an increase in the number of activated CD4\(^+\) and CD8+ lymphocytes in the respective organs with increasing tumor burden. However, mainly CD4\(^+\) T cells, which could constitute both T helper as well as immunosuppressive regulatory T cells, but not CD8\(^+\) T cells, expressed activation markers upon in vitro stimulation when obtained from tumor-bearing mice. Interestingly, these cells from tumor-burdened animals were also functionally hampered in their proliferative response even when subjected to strong in vitro stimulation. Further analyses revealed that the increased frequency of regulatory T cells in tumor-bearing mice is an early event present in all lymphoid organs. Additionally, expression of the immunosuppressive cytokines TGF-β1 and IL-10 became more evident with increased tumor burden. Notably, TGF-β1 is strongly expressed in both the tumor and the tumor-draining lymph node, whereas IL-10 expression is more pronounced in the lymph node, suggesting a more complex regulation of IL-10. Thus, similar to the situation in melanoma patients, both cytokines as well as cellular immune escape mechanisms seem to contribute to the observed immunosuppressed state of tumor-bearing grm1-transgenic mice, suggesting that this model is suitable for preclinical testing of immunomodulatory therapeutics. 2012 2239-2249 Cancer Immunology, Immunotherapy 61 12 urn:nbn:de:bvb:20-opus-125096 10.1007/s00262-012-1290-9 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-5824 Dissertation Alcantarino Menescal, Luciana In vivo characterization of genetic factors involved in Xmrk driven melanoma formation in Medaka (Oryzias latipes): a closer look at braf, Stat5 and c-myc Melanoma arises from the malignant transformation of melanocytes and is one of the most aggressive forms of human cancer. In fish of the genus Xiphophorus, melanoma development, although very rarely, happens spontaneously in nature and can be induced by interspecific crossing. The oncogenic receptor tyrosine kinase, Xmrk, is responsible for melanoma formation in these fishes. Since Xiphophorus are live-bearing fishes and therefore not compatible with embryonic manipulation and transgenesis, the Xmrk melanoma model was brought to the medaka (Oryzias latipes) system. Xmrk expression under the control of the pigment cell specific mitf promoter leads to melanoma formation with 100% penetrance in medaka. Xmrk is an orthologue of the human epidermal growth factor receptor (EGFR) and activates several downstream signaling pathways. Examples of these pathways are the direct phosphorylation of BRAF and Stat5, as well as the enhanced transcription of C-myc. BRAF is a serine-threonine kinase which is found mutated at high frequencies in malignant melanomas. Stat5 is a transcription factor known to be constitutively activated in fish melanoma. C-myc is a transcription factor that is thought to regulate the expression of approximately 15% of all human genes and is involved in cancer progression of a large number of different tumors. To gain new in vivo information on candidate factors known to be involved in melanoma progression, I identified and analysed BRAF, Stat5 and C-myc in the laboratory fish model system medaka. BRAF protein motifs are highly conserved among vertebrates and the results of this work indicate that its function in the MAPK signaling is maintained in medaka. Transgenic medaka lines carrying a constitutive active version of BRAF (V614E) showed more pigmented skin when compared to wild type. Also, some transiently expressing BRAF V614E fishes showed a disrupted eye phenotype. In addition, I was able to identify two Stat5 copies in medaka, named Stat5ab/a and Stat5ab/b. Sequence analysis revealed a higher similarity between both Stat5 sequences when compared to either human Stat5a or Stat5b. This suggests that the two Stat5 copies in medaka arose by an independent duplication processes. I cloned these two Stat5 present in medaka, produced constitutive active and dominant negative gene versions and successfully established transgenic lines carrying each version under the control of the MITF promoter. These lines will help to elucidate questions that are still remaining in Stat5 biology and its function in melanoma progression, like the role of Stat5 phosphorylation on tumor invasiveness. In a third project during my PhD work, I analysed medaka C-myc function and indentified two copies of this gene in medaka, named c-myc17 and c-myc20, according to the chromosome where they are located. I produced conditional transgenic medaka lines carrying the c-myc17 gene coupled to the hormone binding domain of the estrogen receptor to enable specific transgene activation at a given time point. Comparable to human C-myc, medaka C-myc17 is able to induce proliferation and apoptosis in vivo after induction. Besides that, C-myc17 long-term activation led to liver hyperplasia. In summary, the medaka models generated in this work will be important to bring new in vivo information on genes involved in cancer development. Also, the generated transgenic lines can be easily crossed to the melanoma developing Xmrk medaka lines, thereby opening up the possibility to investigate their function in melanoma progression. Besides that, the generated medaka fishes make it possible to follow the whole development of melanocytes, since the embryos are transparent and can be used for high throughput chemical screens. 2012 urn:nbn:de:bvb:20-opus-70762 Graduate School of Life Sciences OPUS4-19696 Wissenschaftlicher Artikel Anelli, Viviana; Ordas, Anita; Kneitz, Susanne; Sagredo, Leonel Munoz; Gourain, Victor; Schartl, Manfred; Meijer, Annemarie H.; Mione, Marina Ras-Induced miR-146a and 193a Target Jmjd6 to Regulate Melanoma Progression Ras genes are among the most commonly mutated genes in human cancer; yet our understanding of their oncogenic activity at the molecular mechanistic level is incomplete. To identify downstream events that mediate ras-induced cellular transformation in vivo, we analyzed global microRNA expression in three different models of Ras-induction and tumor formation in zebrafish. Six microRNAs were found increased in Ras-induced melanoma, glioma and in an inducible model of ubiquitous Ras expression. The upregulation of the microRNAs depended on the activation of the ERK and AKT pathways and to a lesser extent, on mTOR signaling. Two Ras-induced microRNAs (miR-146a and 193a) target Jmjd6, inducing downregulation of its mRNA and protein levels at the onset of Ras expression during melanoma development. However, at later stages of melanoma progression, jmjd6 levels were found elevated. The dynamic of Jmjd6 levels during progression of melanoma in the zebrafish model suggests that upregulation of the microRNAs targeting Jmjd6 may be part of an anti-cancer response. Indeed, triple transgenic fish engineered to express a microRNA-resistant Jmjd6 from the onset of melanoma have increased tumor burden, higher infiltration of leukocytes and shorter melanoma-free survival. Increased JMJD6 expression is found in several human cancers, including melanoma, suggesting that the up-regulation of Jmjd6 is a critical event in tumor progression. The following link has been created to allow review of record GSE37015: http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=jjcrbiuicyyqgpc&acc=GSE37015. 2018 Frontiers in Genetics 9 675 urn:nbn:de:bvb:20-opus-196963 10.3389/fgene.2018.00675 Theodor-Boveri-Institut für Biowissenschaften OPUS4-29042 Wissenschaftlicher Artikel Banicka, Veronika; Martens, Marie Christine; Panzer, Rüdiger; Schrama, David; Emmert, Steffen; Boeckmann, Lars; Thiem, Alexander Homozygous CRISPR/Cas9 knockout generated a novel functionally active exon 1 skipping XPA variant in melanoma cells Defects in DNA repair pathways have been associated with an improved response to immune checkpoint inhibition (ICI). In particular, patients with the nucleotide excision repair (NER) defect disease Xeroderma pigmentosum (XP) responded impressively well to ICI treatment. Recently, in melanoma patients, pretherapeutic XP gene expression was predictive for anti-programmed cell death-1 (PD-1) ICI response. The underlying mechanisms of this finding are still to be revealed. Therefore, we used CRISPR/Cas9 to disrupt XPA in A375 melanoma cells. The resulting subclonal cell lines were investigated by Sanger sequencing. Based on their genetic sequence, candidates from XPA exon 1 and 2 were selected and further analyzed by immunoblotting, immunofluorescence, HCR and MTT assays. In XPA exon 1, we established a homozygous (c.19delG; p.A7Lfs*8) and a compound heterozygous (c.19delG/c.19_20insG; p.A7Lfs*8/p.A7Gfs*55) cell line. In XPA exon 2, we generated a compound heterozygous mutated cell line (c.206_208delTTG/c.208_209delGA; p.I69_D70delinsN/p.D70Hfs*31). The better performance of the homozygous than the heterozygous mutated exon 1 cells in DNA damage repair (HCR) and post-UV-C cell survival (MTT), was associated with the expression of a novel XPA protein variant. The results of our study serve as the fundamental basis for the investigation of the immunological consequences of XPA disruption in melanoma. 2022 International Journal of Molecular Sciences 23 19 urn:nbn:de:bvb:20-opus-290427 10.3390/ijms231911649 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-12621 Wissenschaftlicher Artikel Becker, Jürgen C.; Andersen, Mads H.; Hofmeister-Müller, Valeska; Wobser, Marion; Frey, Lidia; Sandig, Christiane; Walter, Steffen; Singh-Jasuja, Harpreet; Kämpgen, Eckhart; Opitz, Andreas; Zapatka, Marc; Bröcker, Eva-B.; thor Straten, Per; Schrama, David; Ugurel, Selma Survivin-specific T-cell reactivity correlates with tumor response and patient survival: a phase-II peptide vaccination trial in metastatic melanoma Background Therapeutic vaccination directed to induce an anti-tumoral T-cell response is a field of extensive investigation in the treatment of melanoma. However, many vaccination trials in melanoma failed to demonstrate a correlation between the vaccine-specific immune response and therapy outcome. This has been mainly attributed to immune escape by antigen loss, rendering us in the need of new vaccination targets. Patients and methods This phase-II trial investigated a peptide vaccination against survivin, an oncogenic inhibitor-of-apoptosis protein crucial for the survival of tumor cells, in HLA-A1/-A2/-B35-positive patients with treatment-refractory stage-IV metastatic melanoma. The study endpoints were survivin-specific T-cell reactivity (SSTR), safety, response, and survival (OS). Results Sixty-one patients (ITT) received vaccination therapy using three different regimens. 55 patients (PP) were evaluable for response and survival, and 41/55 for SSTR. Patients achieving progression arrest (CR + PR + SD) more often showed SSTRs than patients with disease progression (p = 0.0008). Patients presenting SSTRs revealed a prolonged OS (median 19.6 vs. 8.6 months; p = 0.0077); multivariate analysis demonstrated SSTR as an independent predictor of survival (p = 0.013). The induction of SSTRs was associated with gender (female vs. male; p = 0.014) and disease stage (M1a/b vs. M1c; p = 0.010), but not with patient age, HLA type, performance status, or vaccination regimen. Conclusion Survivin-specific T-cell reactivities strongly correlate with tumor response and patient survival, indicating that vaccination with survivin-derived peptides is a promising treatment strategy in melanoma. 2012 2091-2103 Cancer Immunology, Immunotherapy 61 11 urn:nbn:de:bvb:20-opus-126215 10.1007/s00262-012-1266-9 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-12483 Wissenschaftlicher Artikel Becker, Jürgen C.; Andersen, Mads H.; Hofmeister-Müller, Valeska; Wobser, Marion; Frey, Lidia; Sandig, Christiane; Walter, Steffen; Singh-Jasuja, Harpreet; Kämpgen, Eckhart; Opitz, Andreas; Zapatka, Marc; Bröcker, Eva-B.; thor Straten, Per; Schrama, David; Ugurel, Selma Survivin-specific T-cell reactivity correlates with tumor response and patient survival: a phase-II peptide vaccination trial in metastatic melanoma Background Therapeutic vaccination directed to induce an anti-tumoral T-cell response is a field of extensive investigation in the treatment of melanoma. However, many vaccination trials in melanoma failed to demonstrate a correlation between the vaccine-specific immune response and therapy outcome. This has been mainly attributed to immune escape by antigen loss, rendering us in the need of new vaccination targets. Patients and methods This phase-II trial investigated a peptide vaccination against survivin, an oncogenic inhibitor-of-apoptosis protein crucial for the survival of tumor cells, in HLA-A1/-A2/-B35-positive patients with treatment-refractory stage-IV metastatic melanoma. The study endpoints were survivin-specific T-cell reactivity (SSTR), safety, response, and survival (OS). Results Sixty-one patients (ITT) received vaccination therapy using three different regimens. 55 patients (PP) were evaluable for response and survival, and 41/55 for SSTR. Patients achieving progression arrest (CR + PR + SD) more often showed SSTRs than patients with disease progression (p = 0.0008). Patients presenting SSTRs revealed a prolonged OS (median 19.6 vs. 8.6 months; p = 0.0077); multivariate analysis demonstrated SSTR as an independent predictor of survival (p = 0.013). The induction of SSTRs was associated with gender (female vs. male; p = 0.014) and disease stage (M1a/b vs. M1c; p = 0.010), but not with patient age, HLA type, performance status, or vaccination regimen. Conclusion Survivin-specific T-cell reactivities strongly correlate with tumor response and patient survival, indicating that vaccination with survivin-derived peptides is a promising treatment strategy in melanoma. 2012 2091-2103 Cancer Immunology, Immunotherapy 61 11 urn:nbn:de:bvb:20-opus-124830 10.1007/s00262-012-1266-9 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-26219 Wissenschaftlicher Artikel Esnault, Clara; Schrama, David; Houben, Roland; Guyétant, Serge; Desgranges, Audrey; Martin, Camille; Berthon, Patricia; Viaud-Massuard, Marie-Claude; Touzé, Antoine; Kervarrec, Thibault; Samimi, Mahtab Antibody-drug conjugates as an emerging therapy in oncodermatology Antibody-drug conjugates (ADCs) are an emerging class of therapeutics, with twelve FDA- and EMA-approved drugs for hematological and solid cancers. Such drugs consist in a monoclonal antibody linked to a cytotoxic agent, allowing a specific cytotoxicity to tumor cells. In recent years, tremendous progress has been observed in therapeutic approaches for advanced skin cancer patients. In this regard, targeted therapies (e.g., kinase inhibitors) or immune checkpoint-blocking antibodies outperformed conventional chemotherapy, with proven benefit to survival. Nevertheless, primary and acquired resistances as well as adverse events remain limitations of these therapies. Therefore, ADCs appear as an emerging therapeutic option in oncodermatology. After providing an overview of ADC design and development, the goal of this article is to review the potential ADC indications in the field of oncodermatology. 2022 Cancers 14 3 urn:nbn:de:bvb:20-opus-262192 10.3390/cancers14030778 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-23019 Wissenschaftlicher Artikel Glutsch, Valerie; Amaral, Teresa; Garbe, Claus; Thoms, Kai-Martin; Mohr, Peter; Hauschild, Axel; Schilling, Bastian Indirect Comparison of Combined BRAF and MEK Inhibition in Melanoma Patients with Elevated Baseline Lactate Dehydrogenase The approval of BRAF and MEK inhibitors has signifi-cantly improved treatment outcomes for patients with BRAF-mutated metastatic melanoma. The 3 first-line targeted therapy trials have provided similar results, and thus the identification of predictive biomarkers may generate a more precise basis for clinical deci-sion-making. Elevated baseline lactate dehydrogenase (LDH) has already been determined as a strong prog-nostic factor. Therefore, this indirect analysis compa-red subgroups with elevated baseline LDH across the pivotal targeted therapy trials co-BRIM, COMBI-v and COLUMBUS part 1. The Bucher method was used to compare progression-free survival, objective response rate and overall survival indirectly. The results show a non-significant risk reduction for progression in the subgroup with elevated baseline LDH receiving vemu-rafenib plus cobimetinib compared with dabrafenib plus trametinib and encorafenib plus binimetinib. Al-though an indirect comparison, these data might pro-vide some guidance for treatment recommendations in melanoma patients with elevated LDH. 2020 Acta Dermato-Venereologica 100 urn:nbn:de:bvb:20-opus-230190 10.2340/00015555-3526 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-17726 Wissenschaftlicher Artikel Grimm, Johannes; Hufnagel, Anita; Wobser, Marion; Borst, Andreas; Haferkamp, Sebastian; Houben, Roland; Meierjohann, Svenja BRAF inhibition causes resilience of melanoma cell lines by inducing the secretion of FGF1 Approximately half of all melanoma patients harbour activating mutations in the serine/threonine kinase BRAF. This is the basis for one of the main treatment strategies for this tumor type, the targeted therapy with BRAF and MEK inhibitors. While the initial responsiveness to these drugs is high, resistance develops after several months, frequently at sites of the previously responding tumor. This indicates that tumor response is incomplete and that a certain tumor fraction survives even in drug-sensitive patients, e.g., in a therapy-induced senescence-like state. Here, we show in several melanoma cell lines that BRAF inhibition induces a secretome with stimulating effect on fibroblasts and naive melanoma cells. Several senescence-associated factors were found to be transcribed and secreted in response to BRAF or MEK inhibition, among them members of the fibroblast growth factor family. We identified the growth factor FGF1 as mediator of resilience towards BRAF inhibition, which limits the pro-apoptotic effects of the drug and activates fibroblasts to secrete HGF. FGF1 regulation was mediated by the PI3K pathway and by FRA1, a direct target gene of the MAPK pathway. When FGFR inhibitors were applied in parallel to BRAF inhibitors, resilience was broken, thus providing a rationale for combined therapeutical application. 2018 Oncogenesis 7 71 urn:nbn:de:bvb:20-opus-177261 10.1038/s41389-018-0082-2 Theodor-Boveri-Institut für Biowissenschaften OPUS4-13139 Wissenschaftlicher Artikel Hafner, Christian; Houben, Roland; Baeurle, Anne; Ritter, Cathrin; Schrama, David; Landthaler, Michael; Becker, Jürgen C. Activation of the PI3K/AKT Pathway in Merkel Cell Carcinoma Merkel cell carcinoma (MCC) is a highly aggressive skin cancer with an increasing incidence. The understanding of the molecular carcinogenesis of MCC is limited. Here, we scrutinized the PI3K/AKT pathway, one of the major pathways activated in human cancer, in MCC. Immunohistochemical analysis of 41 tumor tissues and 9 MCC cell lines revealed high levels of AKT phosphorylation at threonine 308 in 88% of samples. Notably, the AKT phosphorylation was not correlated with the presence or absence of the Merkel cell polyoma virus (MCV). Accordingly, knock-down of the large and small T antigen by shRNA in MCV positive MCC cells did not affect phosphorylation of AKT. We also analyzed 46 MCC samples for activating PIK3CA and AKT1 mutations. Oncogenic PIK3CA mutations were found in 2/46 (4%) MCCs whereas mutations in exon 4 of AKT1 were absent. MCC cell lines demonstrated a high sensitivity towards the PI3K inhibitor LY-294002. This finding together with our observation that the PI3K/AKT pathway is activated in the majority of human MCCs identifies PI3K/AKT as a potential new therapeutic target for MCC patients. 2012 e31255 PLoS One 7 2 urn:nbn:de:bvb:20-opus-131398 10.1371/journal.pone.0031255 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-14240 Wissenschaftlicher Artikel Hamm, Henning; Höger, Peter H Skin Tumors in Childhood Background: Dermatologists, paediatricians, and general practitioners are often consulted by worried parents for the evaluation of a cutaneous tumor. Methods: Selective literature review. Results: Only 1-2% of skin tumors excised in children turn out to be malignant when examined histologically. Warning signs of malignancy include rapid growth, firm consistency, diameter exceeding 3 cm, ulceration, a non-movable mass, and presence in the neonatal period. The more common malignant skin tumors in adults-basal cell carcinoma, cutaneous squamous cell carcinoma, and melanoma-are very rare in childhood. Congenital melanocytic nevi and sebaceous nevi bear a lower malignant potential than previously believed; nevertheless, their excision is often indicated. A Spitz nevus can mimic a melanoma both clinically and histologically. Some benign skin tumors of childhood tend to regress spontaneously within a few years but may cause complications at particular locations and when multiple. For infantile hemangiomas requiring systemic treatment because of imminent obstruction or ulceration, propranolol seems to have a far more favorable risk-benefit ratio than corticosteroids. Conclusion: Physicians need specialized knowledge in order to decide whether a skin tumor in a child should be excised, non-surgically treated, or further evaluated, or whether it can be safely left untreated because of the likelihood of spontaneous remission. 2011 347-353 Deutsches Ärzteblatt International 108 20 urn:nbn:de:bvb:20-opus-142402 10.3238/arztebl.2011.0347 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-13710 Wissenschaftlicher Artikel Harter, Patrick N.; Bernatz, Simon; Scholz, Alexander; Zeiner, Pia S.; Zinke, Jenny; Kiyose, Makoto; Blasel, Stella; Beschorner, Rudi; Senft, Christian; Bender, Benjamin; Ronellenfitsch, Michael W.; Wikman, Harriet; Glatzel, Markus; Meinhardt, Matthias; Juratli, Tareq A.; Steinbach, Joachim P.; Plate, Karl H.; Wischhusen, Jörg; Weide, Benjamin; Mittelbronn, Michel Distribution and prognostic relevance of tumor-infiltrating lymphocytes (TILs) and PD-1/PD-L1 immune checkpoints in human brain metastases The activation of immune cells by targeting checkpoint inhibitors showed promising results with increased patient survival in distinct primary cancers. Since only limited data exist for human brain metastases, we aimed at characterizing tumor infiltrating lymphocytes (TILs) and expression of immune checkpoints in the respective tumors. Two brain metastases cohorts, a mixed entity cohort (n = 252) and a breast carcinoma validation cohort (n = 96) were analyzed for CD3+, CD8+, FOXP3+, PD-1+ lymphocytes and PD-L1+ tumor cells by immunohistochemistry. Analyses for association with clinico-epidemiological and neuroradiological parameters such as patient survival or tumor size were performed. TILs infiltrated brain metastases in three different patterns (stromal, peritumoral, diffuse). While carcinomas often show a strong stromal infiltration, TILs in melanomas often diffusely infiltrate the tumors. Highest levels of CD3+ and CD8+ lymphocytes were seen in renal cell carcinomas (RCC) and strongest PD-1 levels on RCCs and melanomas. High amounts of TILs, high ratios of PD-1+/CD8+ cells and high levels of PD-L1 were negatively correlated with brain metastases size, indicating that in smaller brain metastases CD8+ immune response might get blocked. PD-L1 expression strongly correlated with TILs and FOXP3 expression. No significant association of patient survival with TILs was observed, while high levels of PD-L1 showed a strong trend towards better survival in melanoma brain metastases (Log-Rank p = 0.0537). In summary, melanomas and RCCs seem to be the most immunogenic entities. Differences in immunotherapeutic response between tumor entities regarding brain metastases might be attributable to this finding and need further investigation in larger patient cohorts. 2015 13 Oncotarget 6 38 40836 40849 urn:nbn:de:bvb:20-opus-137107 10.18632/oncotarget.5696 Frauenklinik und Poliklinik OPUS4-7217 Dissertation Haydn, Johannes Regulation of ERK1/2 signaling in melanoma Die Mechanismen in einer Zelle, die die Genexpression und somit den Stoffwechsel, das Wachstum und das gesamte Zellverhalten steuern, sind ebenso bedeutsam für das Verständnis der grundlegenden Biologie einer lebenden Zelle wie für die Vorgänge der Krebsentstehung. Dabei bilden hochvernetzte, und strikt regulierte Signaltransduktionswege die Basis für ein belastbares und zugleich hochflexibles regulatorisches Netzwerk. Die Störung solcher Signalkaskaden kann zum einen ursächlich aber auch modifizierend auf die Bildung von Tumoren wirken. Die von Rezeptortyrosinkinasen (RTK) und RAS abhängigen Signalwege, die zur Aktivierung von AKT und ERK1/2 führen, sind hierbei von besonderem Interesse für die Entstehung des malignen Melanoms. Mutationen in Komponenten dieser Wege (z.B. NRAS, BRAF oder PTEN), die die Signalstärke erhöhen kommen in Melanomen sehr häufig vor. Im ersten Teil dieser Arbeit wurden die unterschiedlichen und vielfältigen Funktionen von MKP2, einem Feedbackregulator des ERK1/2-Weges, unter verschiedenen zellulären Rahmenbedingungen, untersucht. Des Weiteren wird eine Funktion des zum AP1-Komplex gehörenden FOSL1, einem unter transkriptioneller Kontrolle des ERK1/2-Weges stehendem Transkriptionsfaktors, hinsichtlich der Steuerung der Zell-Proliferation gezeigt. Weiterhin habe ich Aspekte der direkten pharmakologischen Inhibition des ERK1/2-Weges hinsichtlich ihres Effekts auf die Auslösung von Apoptose untersucht. Aufgrund der Häufigkeit von Mutationen in Genen, die für Proteine des ERK1/2-Weges kodieren (z.B. NRASQ61K, BRAFV600E), gilt die Inhibition dieses Signalwegs als vielversprechende Strategie zur Behandlung des Melanoms. Auch wenn klinische Studien, die Inhibitoren für MEK oder RAF als Einzelmedikamente verwenden, bei mehrmonatiger Behandlung sehr erfolgreich sind, konnten so keine langfristigen Erfolge erzielt werden. Aus diesem Grund werden nun Kombinationstherapien, die einen Inhibitor des ERK1/2-Weges und eine weitere Form der Therapie kombinieren, untersucht. Der zweite Teil dieser Arbeit beschreibt, dass der spezifische MEK Inhibitor PD184352 Melanomzellen vor der Apoptosewirkung von Cisplatin schützen kann. Einzelbehandlung mit Cisplatin führt hierbei zur Akkumulation von DNA Schäden, die wiederum Caspase-abhängig Apoptose induzieren. Zusätzliche Anwendung des MEK Inhibitors verringerte jedoch in einigen Zelllinien das Potential von Cisplatin, Apoptose auszulösen. Diese Zellen zeigten eine verstärkte Aktivierung der Serin/Threonin-KInase AKT nach MEK Inhibition. Diese AKT Aktivierung führte zur Inaktivierung der FOXO Transkriptionsfaktoren, was wiederum die Expression des pro-apoptotischen BH3-only Proteins PUMA verringerte. PUMA selbst ist ein wichtiger Bestandteil der Apoptose Maschinerie, die durch Cisplatin aktiviert wird. Die im Rahmen dieser Arbeit erhaltenen Befunde deuten darauf hin, dass RTKs, im besonderen EGFR, bei diesem Crosstalk eine Rolle spielen. Diese Ergebnisse zeigen, dass die Inhibition des RAS/RAF/MEK/ERK Signalweges im Melanom nicht zwangsläufig von Vorteil sein muss, falls die Zellen gleichzeitig mit einem genotoxischen Medikament behandelt werden. Hier kann sie sogar die Überlebensfähigkeit von Melanomzellen unter Apoptose induzierenden Bedingungen verbessern. 2012 urn:nbn:de:bvb:20-opus-85727 Theodor-Boveri-Institut für Biowissenschaften OPUS4-12064 Wissenschaftlicher Artikel Haydn, Johannes M.; Hufnagel, Anita; Grimm, Johannes; Maurus, Katja; Schartl, Manfred; Meierjohann, Svenja The MAPK pathway as an apoptosis enhancer in melanoma Inhibition of RAF/MEK/ERK signaling is beneficial for many patients with BRAFV600E-mutated melanoma. However, primary and secondary resistances restrict long-lasting therapy success. Combination therapies are therefore urgently needed. Here, we evaluate the cellular effect of combining a MEK inhibitor with a genotoxic apoptosis inducer. Strikingly, we observed that an activated MAPK pathway promotes in several melanoma cell lines the pro-apoptotic response to genotoxic stress, and MEK inhibition reduces intrinsic apoptosis. This goes along with MEK inhibitor induced increased RAS and P-AKT levels. The protective effect of the MEK inhibitor depends on PI3K signaling, which prevents the induction of pro-apoptotic PUMA that mediates apoptosis after DNA damage. We could show that the MEK inhibitor dependent feedback loop is enabled by several factors, including EGF receptor and members of the SPRED family. The simultaneous knockdown of SPRED1 and SPRED2 mimicked the effects of MEK inhibitor such as PUMA repression and protection from apoptosis. Our data demonstrate that MEK inhibition of BRAFV600E-positive melanoma cells can protect from genotoxic stress, thereby achieving the opposite of the intended anti-tumorigenic effect of the combination of MEK inhibitor with inducers of intrinsic apoptosis. 2014 5040-53 Oncotarget 5 13 urn:nbn:de:bvb:20-opus-120649 Theodor-Boveri-Institut für Biowissenschaften OPUS4-26061 Wissenschaftlicher Artikel Helmprobst, Frederik; Kneitz, Susanne; Klotz, Barbara; Naville, Magali; Dechaud, Corentin; Volff, Jean-Nicolas; Schartl, Manfred Differential expression of transposable elements in the medaka melanoma model Malignant melanoma incidence is rising worldwide. Its treatment in an advanced state is difficult, and the prognosis of this severe disease is still very poor. One major source of these difficulties is the high rate of metastasis and increased genomic instability leading to a high mutation rate and the development of resistance against therapeutic approaches. Here we investigate as one source of genomic instability the contribution of activation of transposable elements (TEs) within the tumor. We used the well-established medaka melanoma model and RNA-sequencing to investigate the differential expression of TEs in wildtype and transgenic fish carrying melanoma. We constructed a medaka-specific TE sequence library and identified TE sequences that were specifically upregulated in tumors. Validation by qRT- PCR confirmed a specific upregulation of a LINE and an LTR element in malignant melanomas of transgenic fish. 2021 PLoS One 16 10 urn:nbn:de:bvb:20-opus-260615 10.1371/journal.pone.0251713 Theodor-Boveri-Institut für Biowissenschaften OPUS4-17364 Wissenschaftlicher Artikel Heuser, Christoph; Gotot, Janine; Piotrowski, Eveline Christina; Philipp, Marie-Sophie; Courrèges, Christina Johanna Felicia; Otte, Martin Sylvester; Guo, Linlin; Schmid-Burgk, Jonathan Leo; Hornung, Veit; Heine, Annkristin; Knolle, Percy Alexander; Garbi, Natalio; Serfling, Edgar; Evaristo, César; Thaiss, Friedrich; Kurts, Christian Prolonged IKK\(\beta\) Inhibition Improves Ongoing CTL Antitumor Responses by Incapacitating Regulatory T Cells Regulatory T cells (Tregs) prevent autoimmunity but limit antitumor immunity. The canonical NF-\(\kappa\)B signaling pathway both activates immunity and promotes thymic Treg development. Here, we report that mature Tregs continue to require NF-\(\kappa\)B signaling through I\(\kappa\)B-kinase \(\beta\) (IKK\(\beta\)) after thymic egress. Mice lacking IKK\(\beta\) in mature Tregs developed scurfy-like immunopathology due to death of peripheral FoxP3\(^+\) Tregs. Also, pharmacological IKK\(\beta\) inhibition reduced Treg numbers in the circulation by ~50% and downregulated FoxP3 and CD25 expression and STAT5 phosphorylation. In contrast, activated cytotoxic T lymphocytes (CTLs) were resistant to IKK\(\beta\) inhibition because other pathways, in particular nuclear factor of activated T cells (NFATc1) signaling, sustained their survival and expansion. In a melanoma mouse model, IKK\(\beta\) inhibition after CTL cross-priming improved the antitumor response and delayed tumor growth. In conclusion, prolonged IKK\(\beta\) inhibition decimates circulating Tregs and improves CTL responses when commenced after tumor vaccination, indicating that IKK\(\beta\) represents a druggable checkpoint. 2017 578-586 Cell Reports 21 3 urn:nbn:de:bvb:20-opus-173643 10.1016/j.celrep.2017.09.082 Pathologisches Institut OPUS4-23892 Wissenschaftlicher Artikel Hoesl, Christine; Fröhlich, Thomas; Posch, Christian; Kneitz, Hermann; Goebeler, Matthias; Schneider, Marlon R.; Dahlhoff, Maik The transmembrane protein LRIG1 triggers melanocytic tumor development following chemically induced skin carcinogenesis The incidence of melanoma and nonmelanoma skin cancer has increased tremendously in recent years. Although novel treatment options have significantly improved patient outcomes, the prognosis for most patients with an advanced disease remains dismal. It is, thus, imperative to understand the molecular mechanisms involved in skin carcinogenesis in order to develop new targeted treatment strategies. Receptor tyrosine kinases (RTK) like the ERBB receptor family, including EGFR/ERBB1, ERBB2/NEU, ERBB3, and ERBB4, are important regulators of skin homeostasis and their dysregulation often results in cancer, which makes them attractive therapeutic targets. Members of the leucine-rich repeats and immunoglobulin-like domains protein family (LRIG1-3) are ERBB regulators and thus potential therapeutic targets to manipulate ERBB receptors. Here, we analyzed the function of LRIG1 during chemically induced skin carcinogenesis in transgenic mice expressing LRIG1 in the skin under the control of the keratin 5 promoter (LRIG1-TG mice). We observed a significant induction of melanocytic tumor formation in LRIG1-TG mice and no difference in papilloma incidence between LRIG1-TG and control mice. Our findings also revealed that LRIG1 affects ERBB signaling via decreased phosphorylation of EGFR and increased activation of the oncoprotein ERBB2 during skin carcinogenesis. The epidermal proliferation rate was significantly decreased during epidermal tumorigenesis under LRIG1 overexpression, and the apoptosis marker cleaved caspase 3 was significantly activated in the epidermis of transgenic LRIG1 mice. Additionally, we detected LRIG1 expression in human cutaneous squamous cell carcinoma and melanoma samples. Therefore, we depleted LRIG1 in human melanoma cells (A375) by CRISPR/Cas9 technology and found that this caused EGFR and ERBB3 downregulation in A375 LRIG1 knockout cells 6 h following stimulation with EGF. In conclusion, our study demonstrated that LRIG1-TG mice develop melanocytic skin tumors during chemical skin carcinogenesis and a deletion of LRIG1 in human melanoma cells reduces EGFR and ERBB3 expression after EGF stimulation. 2021 15 Molecular Oncology 15 8 2140 2155 urn:nbn:de:bvb:20-opus-238925 10.1002/1878-0261.12945 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-12219 Wissenschaftlicher Artikel Hohenauer, Tobias; Berking, Carola; Schmidt, Andreas; Haferkamp, Sebastian; Senft, Daniela; Kammerbauer, Claudia; Fraschka, Sabine; Graf, Saskia Anna; Irmler, Martin; Beckers, Johannes; Flaig, Michael; Aigner, Achim; Höbel, Sabrina; Hoffmann, Franziska; Hermeking, Heiko; Rothenfusser, Simon; Endres, Stefan; Ruzicka, Thomas; Besch, Robert The neural crest transcription factor Brn3a is expressed in melanoma and required for cell cycle progression and survival Pigment cells and neuronal cells both are derived from the neural crest. Here, we describe the Pit-Oct-Unc (POU) domain transcription factor Brn3a, normally involved in neuronal development, to be frequently expressed in melanoma, but not in melanocytes and nevi. RNAi-mediated silencing of Brn3a strongly reduced the viability of melanoma cell lines and decreased tumour growth in vivo. In melanoma cell lines, inhibition of Brn3a caused DNA double-strand breaks as evidenced by Mre11/Rad50-containing nuclear foci. Activated DNA damage signalling caused stabilization of the tumour suppressor p53, which resulted in cell cycle arrest and apoptosis. When Brn3a was ectopically expressed in primary melanocytes and fibroblasts, anchorage-independent growth was increased. In tumourigenic melanocytes and fibroblasts, Brn3a accelerated tumour growth in vivo. Furthermore, Brn3a cooperated with proliferation pathways such as oncogenic BRAF, by reducing oncogene-induced senescence in non-malignant melanocytes. Together, these results identify Brn3a as a new factor in melanoma that is essential for melanoma cell survival and that promotes melanocytic transformation and tumourigenesis. 2013 919-934 EMBO Molecular Medicine 5 urn:nbn:de:bvb:20-opus-122193 10.1002/emmm.201201862 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-28810 Wissenschaftlicher Artikel Isberner, Nora; Gesierich, Anja; Balakirouchenane, David; Schilling, Bastian; Aghai-Trommeschlaeger, Fatemeh; Zimmermann, Sebastian; Kurlbaum, Max; Puszkiel, Alicja; Blanchet, Benoit; Klinker, Hartwig; Scherf-Clavel, Oliver Monitoring of dabrafenib and trametinib in serum and self-sampled capillary blood in patients with BRAFV600-mutant melanoma Simple Summary In melanoma patients treated with dabrafenib and trametinib, dose reductions and treatment discontinuations related to adverse events (AE) occur frequently. However, the associations between patient characteristics, AE, and exposure are unclear. Our prospective study analyzed serum (hydroxy-)dabrafenib and trametinib exposure and investigated its association with toxicity and patient characteristics. Additionally, the feasibility of at-home sampling of capillary blood was assessed, and a model to convert capillary blood concentrations to serum concentrations was developed. (Hydroxy-)dabrafenib or trametinib exposure was not associated with age, sex, body mass index, or AE. Co-medication with P-glycoprotein inducers was associated with lower trough concentrations of trametinib but not (hydroxy-)dabrafenib. The applicability of the self-sampling of capillary blood was demonstrated. Our conversion model was adequate for estimating serum exposure from micro-samples. The monitoring of dabrafenib and trametinib may be useful for dose modification and can be optimized by at-home sampling and our new conversion model. Abstract Patients treated with dabrafenib and trametinib for BRAF\(^{V600}\)-mutant melanoma often experience dose reductions and treatment discontinuations. Current knowledge about the associations between patient characteristics, adverse events (AE), and exposure is inconclusive. Our study included 27 patients (including 18 patients for micro-sampling). Dabrafenib and trametinib exposure was prospectively analyzed, and the relevant patient characteristics and AE were reported. Their association with the observed concentrations and Bayesian estimates of the pharmacokinetic (PK) parameters of (hydroxy-)dabrafenib and trametinib were investigated. Further, the feasibility of at-home sampling of capillary blood was assessed. A population pharmacokinetic (popPK) model-informed conversion model was developed to derive serum PK parameters from self-sampled capillary blood. Results showed that (hydroxy-)dabrafenib or trametinib exposure was not associated with age, sex, body mass index, or toxicity. Co-medication with P-glycoprotein inducers was associated with significantly lower trough concentrations of trametinib (p = 0.027) but not (hydroxy-)dabrafenib. Self-sampling of capillary blood was feasible for use in routine care. Our conversion model was adequate for estimating serum PK parameters from micro-samples. Findings do not support a general recommendation for monitoring dabrafenib and trametinib but suggest that monitoring can facilitate making decisions about dosage adjustments. To this end, micro-sampling and the newly developed conversion model may be useful for estimating precise PK parameters. 2022 Cancers 14 19 urn:nbn:de:bvb:20-opus-288109 10.3390/cancers14194566 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-14602 Wissenschaftlicher Artikel Kneitz, Susanne; Mishra, Rasmi R.; Chalopin, Domitille; Postlethwait, John; Warren, Wesley C.; Walther, Ronald B.; Schartl, Manfred Germ cell and tumor associated piRNAs in the medaka and \(Xiphophorus\) melanoma models Background A growing number of studies report an abnormal expression of Piwi-interacting RNAs (piRNAs) and the piRNA processing enzyme Piwi in many cancers. Whether this finding is an epiphenomenon of the chaotic molecular biology of the fast dividing, neoplastically transformed cells or is functionally relevant to tumorigenesisis is difficult to discern at present. To better understand the role of piRNAs in cancer development small laboratory fish models can make a valuable contribution. However, little is known about piRNAs in somatic and neoplastic tissues of fish. Results To identify piRNA clusters that might be involved in melanoma pathogenesis, we use several transgenic lines of medaka, and platyfish/swordtail hybrids, which develop various types of melanoma. In these tumors Piwi, is expressed at different levels, depending on tumor type. To quantify piRNA levels, whole piRNA populations of testes and melanomas of different histotypes were sequenced. Because no reference piRNA cluster set for medaka or Xiphophorus was yet available we developed a software pipeline to detect piRNA clusters in our samples and clusters were selected that were enriched in one or more samples. We found several loci to be overexpressed or down-regulated in different melanoma subtypes as compared to hyperpigmented skin. Furthermore, cluster analysis revealed a clear distinction between testes, low-grade and high-grade malignant melanoma in medaka. Conclusions Our data imply that dysregulation of piRNA expression may be associated with development of melanoma. Our results also reinforce the importance of fish as a suitable model system to study the role of piRNAs in tumorigenesis. 2016 BMC Genomics 17 357 urn:nbn:de:bvb:20-opus-146028 10.1186/s12864-016-2697-z Theodor-Boveri-Institut für Biowissenschaften OPUS4-21120 Wissenschaftlicher Artikel Kosnopfel, Corinna; Sinnberg, Tobias; Sauer, Birgit; Niessner, Heike; Muenchow, Alina; Fehrenbacher, Birgit; Schaller, Martin; Mertens, Peter R.; Garbe, Claus; Thakur, Basant Kumar; Schittek, Birgit Tumour progression stage-dependent secretion of YB-1 stimulates melanoma cell migration and invasion Secreted factors play an important role in intercellular communication. Therefore, they are not only indispensable for the regulation of various physiological processes but can also decisively advance the development and progression of tumours. In the context of inflammatory disease, Y-box binding protein 1 (YB-1) is actively secreted and the extracellular protein promotes cell proliferation and migration. In malignant melanoma, intracellular YB-1 expression increases during melanoma progression and represents an unfavourable prognostic marker. Here, we show active secretion of YB-1 from melanoma cells as opposed to benign cells of the skin. Intriguingly, YB-1 secretion correlates with the stage of melanoma progression and depends on a calcium- and ATP-dependent non-classical secretory pathway leading to the occurrence of YB-1 in the extracellular space as a free protein. Along with an elevated YB-1 secretion of melanoma cells in the metastatic growth phase, extracellular YB-1 exerts a stimulating effect on melanoma cell migration, invasion, and tumourigenicity. Collectively, these data suggest that secreted YB-1 plays a functional role in melanoma cell biology, stimulating metastasis, and may serve as a novel biomarker in malignant melanoma that reflects tumour aggressiveness. 2020 Cancers 12 8 urn:nbn:de:bvb:20-opus-211206 10.3390/cancers12082328 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-4253 Dissertation Laisney, Juliette Agnès Geneviève Claire Characterisation and regulation of the Egfr/Egfr ligand system in fish models for melanoma Fish of the genus Xiphophorus belong to the oldest animal models in cancer research. The oncogene responsible for the generation of spontaneous aggressive melanoma encodes for a mutated epidermal growth factor receptor (Egfr) and is called xmrk for Xiphophorus melanoma receptor kinase. Xmrk constitutive activation mechanisms and subsequent signaling pathways have already been investigated and charaterized but it is still unknown if Egfr ligands may also play a role in Xmrk-driven melanoma formation. To investigate the potential role of Egfr ligands in Xmrk-driven melanoma, I firstly analyzed the evolution of teleost and tetrapod Egfr/Egfr ligand systems. I especially focused on the analysis on the medaka fish, a closely related species to Xiphophorus, for which the whole genome has been sequenced. I could identify all seven Egfr ligands in medaka and could show that the two teleost-specific Egfr copies of medaka display dissimilar expression patterns in adult tissues together with differential expression of Egfr ligand subsets, arguing for subfunctionalization of receptor functions in this fish. Our phylogenetic and synteny analyses supported the hypothesis that only one gene in the chordate ancestor gave rise to the diversity of Egfr ligands found in vertebrate genomes today. I also could show that the Egfr extracellular subdomains implicated in ligand binding are not evolutionary conserved between tetrapods and teleosts, making the use of heterologous ligands in experiments with fish cells debatable. Despite its well understood and straight-forward process, Xmrk-driven melanomagenesis in Xiphophorus is problematic to further investigate in vivo. Our laboratory recently established a new melanoma animal model by generating transgenic mitf::xmrk medaka fishes, a Xiphophorus closely related species offering many more advantages. These fishes express xmrk under the control of the pigment-cell specific Mitf promoter. During my PhD thesis, I participated in the molecular analysis of the stably transgenic medaka and could show that the Xmrk-induced signaling pathways are similar when comparing Xiphophorus with transgenic mitf::xmrk medaka. These data together with additional RNA expression, protein, and histology analyses showed that Xmrk expression under the control of a pigment cell-specific promoter is sufficient to induce melanoma in the transgenic medaka, which develop very stereotyped tumors, including uveal and extracutaneous melanoma, with early onset during larval stages. To further investigate the potential role of Egfr ligands in Xmrk-driven melanoma, I made use of two model systems. One of them was the above mentioned mitf::xmrk medaka, the other was an in-vitro cell culture system, where the EGF-inducible Xmrk chimera HERmrk is stably expressed in murine melanocytes. Here I could show that HERmrk activation strongly induced expression of amphiregulin (Areg) and heparin-binding EGF-like growth factor (Hbegf) in melanocytes. This regulation was dependent on the MAPK and SRC signaling pathways. Moreover, upregulation of Adam10 and Adam17, the two major sheddases of Egfr ligands, was observed. I also could demonstrate the functionality of the growth factors by invitro analyses. Using the mitf::xmrk medaka model I could also show the upregulation of a subset of ligand genes, namely egf, areg, betacellulin (btc) and epigen (epgn) as well as upregulation of medaka egfrb in tumors from fish with metastatic melanoma. All these results converge to support an Xmrk-induced autocrine Egfr ligand loop. Interestingly, my in-vitro experiments with conditioned supernatant from medaka Egf- and Hbegf-producing cells revealed that not only Xiphophorus Egfrb, but also the pre-activated Xmrk could be further stimulated by the ligands. Altogether, I could show with in-vitro and in-vivo experiments that Xmrk is capable of inducing a functional autocrine Egfr ligand loop. These data confirm the importance of autocrine loops in receptor tyrosine kinase (RTK)-dependent cancer development and show the possibility for a constitutively active RTK to strengthen its oncogenic signaling by ligand binding. 2010 urn:nbn:de:bvb:20-opus-51369 Theodor-Boveri-Institut für Biowissenschaften OPUS4-6740 Dissertation Leikam, Claudia Oncogene-induced senescence in melanocytes Melanoma is the most aggressive skin cancer with very limited treatment options. Upon appearance of metastases chemotherapeutics are used to either kill or slow down the growth of cancer cells by inducing apoptosis or senescence, respectively. With melanomas originating from melanocytes, it is vital to elucidate the mechanisms that distinguish senescence induction from proliferation and tumourigenicity. Xmrk (Xiphophorus melanoma receptor kinase), the fish orthologue of the human epidermal growth factor receptor (EGFR), causes highly aggressive melanoma in fish. Using an inducible variant, HERmrk, I showed that high receptor levels result in melanocyte senescence, whereas low and medium expression allows for cell proliferation and tumourigenicity. Mechanistically, HERmrk leads to increased reactive oxygen species (ROS) levels, which trigger a DNA damage response. Consequently, multinucleated, senescent cells develop by both endomitosis and fusion. Furthermore, oncogenic N-RAS (N--RAS61K) induces a similar multinucleated phenotype in melanocytes. In addition, I found that both overexpression of C-MYC and the knockdown of miz­-1 (Myc­-interacting zinc finger protein 1) diminished HERmrk-induced senescence entry. C-MYC prevent ROS induction, DNA damage and senescence, while acting synergistically with HERmrk in conveying tumourigenic features to melanocytes. Further analyses identified cystathionase (CTH) as a novel target gene of Myc and Miz-­1 crucial for senescence prevention. CTH encodes an enzyme involved in the synthesis of cysteine from methionine, thereby allowing for increased ROS detoxification. Even though senescence was thought to be irreversible and hence tumour protective, I demonstrated that prolonged expression of the melanoma oncogene N­-RAS61K in pigment cells overcomes initial OIS by triggering the emergence of tumour-initiating, mononucleated stem-like cells from multinucleated senescent cells. This progeny is dedifferentiated, highly proliferative, anoikis­-resistant and induces fast­-growing, metastatic tumours upon transplantation into nude mice. Our data demonstrate that induction of OIS is not only a cellular failsafe mechanism, but also carries the potential to provide a source for highly aggressive, tumour­-initiating cells. 2012 urn:nbn:de:bvb:20-opus-79316 Graduate School of Life Sciences OPUS4-21543 Wissenschaftlicher Artikel Livingstone, E.; Zaremba, A.; Horn, S.; Ugurel, S.; Casalini, B.; Schlaak, M.; Hassel, J.C.; Herbst, R.; Utikal, J.S.; Weide, B.; Gutzmer, R.; Meier, F.; Koelsche, C.; Hadaschik, E.; Sucker, A.; Reis, H.; Merkelbach-Bruse, S.; Siewert, M.; Sahm, F.; von Deimling, A.; Cosgarea, I.; Zimmer, L.; Schadendorf, D.; Schilling, B.; Griewank, K.G. GNAQ and GNA11 mutant nonuveal melanoma: a subtype distinct from both cutaneous and uveal melanoma Background GNAQ and GNA11 mutant nonuveal melanoma represent a poorly characterized rare subgroup of melanoma with a gene mutation profile similar to uveal melanoma. Objectives To characterize these tumours in terms of clinical behaviour and genetic characteristics. Methods Patients with nonuveal GNAQ/11 mutated melanoma were identified from the prospective multicentre tumour tissue registry ADOREG, Tissue Registry in Melanoma (TRIM) and additional cooperating skin cancer centres. Extensive data on patient, tumour and treatment characteristics were collected retrospectively. Targeted sequencing was used to determine tumour mutational burden. Immunohistochemistry staining was performed for programmed death-ligand 1 and BRCA1-associated protein (BAP)1. Existing whole-exome cutaneous and uveal melanoma data were analysed for mutation type and burden. Results We identified 18 patients with metastatic GNAQ/11 mutant nonuveal melanoma. Tumours had a lower tumour mutational burden and fewer ultraviolet signature mutations than cutaneous melanomas. In addition to GNAQ and GNA11 mutations (nine each), six splicing factor 3b subunit 1 (SF3B1), three eukaryotic translation initiation factor 1A X-linked (EIF1AX) and four BAP1 mutations were detected. In contrast to uveal melanoma, GNAQ/11 mutant nonuveal melanomas frequently metastasized lymphatically and concurrent EIF1AX, SF3B1 and BAP1 mutations showed no apparent association with patient prognosis. Objective response to immunotherapy was poor with only one partial response observed in 10 treated patients (10%). Conclusions Our findings suggest that GNAQ/11 mutant nonuveal melanomas are a subtype of melanoma that is both clinically and genetically distinct from cutaneous and uveal melanoma. As they respond poorly to available treatment regimens, novel effective therapeutic approaches for affected patients are urgently needed. What is already known about this topic? The rare occurrence of GNAQ/11 mutations in nonuveal melanoma has been documented. GNAQ/11 mutant nonuveal melanomas also harbour genetic alterations in EIF1AX, SF3B1 and BAP1 that are of prognostic relevance in uveal melanoma. What does this study add? GNAQ/11 mutant nonuveal melanomas show metastatic spread reminiscent of cutaneous melanoma, but not uveal melanoma. GNAQ/11 mutant nonuveal melanomas have a low tumour mutational burden that is higher than uveal melanoma, but lower than cutaneous melanoma. What is the translational message? Primary GNAQ/11 mutant nonuveal melanomas are a subtype of melanoma that is clinically and genetically distinct from both cutaneous and uveal melanoma. As metastatic GNAQ/11 mutant nonuveal melanomas respond poorly to available systemic therapies, including immune checkpoint inhibition, novel therapeutic approaches for these tumours are urgently needed. 2020 11 British Journal of Dermatology 183 5 928 939 urn:nbn:de:bvb:20-opus-215434 10.1111/bjd.18947 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-23958 Wissenschaftlicher Artikel Lodde, Georg; Forschner, Andrea; Hassel, Jessica; Wulfken, Lena M.; Meier, Friedegund; Mohr, Peter; Kähler, Katharina; Schilling, Bastian; Loquai, Carmen; Berking, Carola; Hüning, Svea; Schatton, Kerstin; Gebhardt, Christoffer; Eckardt, Julia; Gutzmer, Ralf; Reinhardt, Lydia; Glutsch, Valerie; Nikfarjam, Ulrike; Erdmann, Michael; Stang, Andreas; Kowall, Bernd; Roesch, Alexander; Ugurel, Selma; Zimmer, Lisa; Schadendorf, Dirk; Livingstone, Elisabeth Factors influencing the adjuvant therapy decision: results of a real-world multicenter data analysis of 904 melanoma patients Adjuvant treatment of melanoma patients with immune-checkpoint inhibition (ICI) and targeted therapy (TT) significantly improved recurrence-free survival. This study investigates the real-world situation of 904 patients from 13 German skin cancer centers with an indication for adjuvant treatment since the approval of adjuvant ICI and TT. From adjusted log-binomial regression models, we estimated relative risks for associations between various influence factors and treatment decisions (adjuvant therapy yes/no, TT vs. ICI in BRAF mutant patients). Of these patients, 76.9% (95% CI 74-80) opted for a systemic adjuvant treatment. The probability of starting an adjuvant treatment was 26% lower in patients >65 years (RR 0.74, 95% CI 68-80). The most common reasons against adjuvant treatment given by patients were age (29.4%, 95% CI 24-38), and fear of adverse events (21.1%, 95% CI 16-28) and impaired quality of life (11.9%, 95% CI 7-16). Of all BRAF-mutated patients who opted for adjuvant treatment, 52.9% (95% CI 47-59) decided for ICI. Treatment decision for TT or ICI was barely associated with age, gender and tumor stage, but with comorbidities and affiliated center. Shortly after their approval, adjuvant treatments have been well accepted by physicians and patients. Age plays a decisive role in the decision for adjuvant treatment, while pre-existing autoimmune disease and regional differences influence the choice between TT or ICI. 2021 Cancers 13 10 urn:nbn:de:bvb:20-opus-239583 10.3390/cancers13102319 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-12558 Wissenschaftlicher Artikel Meierjohann, Svenja Hypoxia independent drivers of melanoma angiogenesis Tumor angiogenesis is a process which is traditionally regarded as the tumor's response to low nutrient supply occurring under hypoxic conditions. However, hypoxia is not a pre-requisite for angiogenesis. The fact that even single tumor cells or small tumor cell aggregates are capable of attracting blood vessels reveals the early metastatic capability of tumor cells. This review sheds light on the hypoxia-independent mechanisms of tumor angiogenesis in melanoma. 2015 Frontiers in Oncology 5 120 urn:nbn:de:bvb:20-opus-125586 10.3389/fonc.2015.00102 Theodor-Boveri-Institut für Biowissenschaften OPUS4-6787 Dissertation Regneri, Janine Transcriptional regulation of cancer genes in the Xiphophorus melanoma system The Xiphophorus melanoma system is a useful animal model for the study of the genetic basis of tumor formation. The development of hereditary melanomas in interspecific hybrids of Xiphophorus is connected to pigment cell specific overexpression of the mutationally activated receptor tyrosine kinase Xmrk. In purebred fish the oncogenic function of xmrk is suppressed by the molecularly still unidentified locus R. The xmrk oncogene was generated by a gene duplication event from the Xiphophorus egfrb gene and thereby has acquired a new 5' regulatory sequence, which has probably altered the transcriptional control of the oncogene. So far, the xmrk promoter region was still poorly characterized and the molecular mechanism by which R controls xmrk-induced melanoma formation in Xiphophorus still remained to be elucidated. To test the hypothesis that R controls melanoma development in Xiphophorus on the transcriptional level, the first aim of the thesis was to gain a deeper insight into the transcriptional regulation of the xmrk oncogene. To this end, a quantitative analysis of xmrk transcript levels in different Xiphophorus genotypes carrying either the highly tumorigenic xmrkB or the non-tumorigenic xmrkA allele was performed. I was able to demonstrate that expression of the tumorigenic xmrkB allele is strongly increased in malignant melanomas of R-free backcross hybrids compared to benign lesions, macromelanophore spots, and healthy skin. The expression level of the non-tumorigenic xmrkA allele, in contrast, is not influenced by the presence or absence of R. These findings strongly indicate that differential transcriptional regulation of the xmrk promoter triggers the tumorigenic potential of these xmrk alleles. To functionally characterize the xmrk promoter region, I established a luciferase assay using BAC clones containing the genomic regions where xmrk and egfrb are located for generation of reporter constructs. This approach showed for the first time a melanoma cell specific transcriptional activation of xmrkB by its flanking regions, thereby providing the first functional evidence that the xmrk oncogene is controlled by a pigment cell specific promoter region. Subsequent analysis of different deletion constructs of the xmrkB BAC reporter construct strongly indicated that the regulatory elements responsible for the tumor-inducing overexpression of xmrkB in melanoma cells are located within 67 kb upstream of the xmrk oncogene. Taken together, these data indicate that melanoma formation in Xiphophorus is regulated by a tight transcriptional control of the xmrk oncogene and that the R locus acts through this mechanism. As the identification of the R-encoded gene(s) is necessary to fully understand how melanoma formation in Xiphophorus is regulated, I furthermore searched for alternative R candidate genes in this study. To this end, three genes, which are located in the genomic region where R has been mapped, were evaluated for their potential to be a crucial constituent of the regulator locus R. Among these genes, I identified pdcd4a, the ortholog of the human tumor suppressor gene PDCD4, as promising new candidate, because this gene showed the expression pattern expected from the crucial tumor suppressor gene encoded at the R locus. 2013 urn:nbn:de:bvb:20-opus-82319 Theodor-Boveri-Institut für Biowissenschaften OPUS4-2304 Dissertation Robubi, Armin RAF Kinases: Pathway, Modulation and Modeling The Ras/RAF/MEK/ERK cascade is a central cellular signal transduction pathway involved in cell proliferation, differentiation, and survival where RAF kinases are pivotal kinases implicated in cancer. The development of specific irreversible kinase inhibitors is a rewarding but difficult aim. CI-1033 was developed to irreversibly inhibit erbB receptor tyrosine kinases by reacting to the Cys113 residue (p38alpha MAP kinase numbering) of the kinase domain. In this study we tried a similar approach to target the RAF oncoproteins which posses a similar cysteine at position 108 in the hinge region between the small n-lobe and the large c-lobe of the kinase domain. A novel synthetic approach including a lyophilization step allowed us the synthesis of a diphenyl urea compound with an epoxide moiety (compound 1). Compound 1 possessed inhibitory activity in vitro. However our time kinetics experiments and mass spectroscopic studies clearly indicate that compound 1 does not react covalently with the cysteine residue in the hinge region. Moreover, in cell culture experiments, a strong activation of the RAF signaling pathway was observed, an effect which is known from several other RAF kinase inhibitors and is here reported for the first time for a diphenyl urea compound, to which the clinically used unspecific kinase inhibitor BAY 43-9006 (Sorafinib, Nexavar) belongs. Although activation was apparently independent on B- and C-RAF hetero-oligomerization in vitro, in vivo experiments support such a mechanism as the activation did not occur in starved knockout cells lacking either B-RAF or C-RAF. Furthermore, we developed a mathematical model of the Ras/RAF/MEK/ERK cascade demonstrating how stimuli induce different signal patterns and thereby different cellular responses, depending on cell type and the ratio between B-RAF and C-RAF. Based on biochemical data for activation and dephosphorylation, we set up differential equations for a dynamical model of the Ras/RAF/MEK/ERK cascade. We find a different signaling pattern and response result for B-RAF (strong activation, sustained signal) and C-RAF (steep activation, transient signal). We further support the significance of such differential modulatory signaling by showing different RAF isoform expression in various cell lines and experimental testing of the predicted kinase activities in B-RAF, C-RAF as well as mutated versions. Additionally the effect of the tumor suppressor DiRas3 (also known as Noey2 or ARHI) on RAF signaling was studied. I could show that DiRas3 down-regulates the mitogenic pathway by inhibition of MEK, a basis for a refined model of the Ras/RAF/MEK/ERK cascade. 2007 urn:nbn:de:bvb:20-opus-26953 Theodor-Boveri-Institut für Biowissenschaften OPUS4-17181 Wissenschaftlicher Artikel Rothe, Hansjörg; Brandenburg, Vincent; Haun, Margot; Kollerits, Barbara; Kronenberg, Florian; Ketteler, Markus; Wanner, Christoph Ecto-5 ' -Nucleotidase CD73 (NT5E), vitamin D receptor and FGF23 gene polymorphisms may play a role in the development of calcific uremic arteriolopathy in dialysis patients - Data from the German Calciphylaxis Registry Introduction: Calciphylaxis/calcific uremic arteriolopathy affects mainly end-stage kidney disease patients but is also associated with malignant disorders such as myeloma, melanoma and breast cancer. Genetic risk factors of calciphylaxis have never been studied before. Methods: We investigated 10 target genes using a tagging SNP approach: the genes encoding CD73/ ecto-5'-nucleotidase (purinergic pathway), Matrix Gla protein, Fetuin A, Bone Gla protein, VKORC1 (all related to intrinsic calcification inhibition), calcium-sensing receptor, FGF23, Klotho, vitamin D receptor, stanniocalcin 1 (all related to CKD-MBD). 144 dialysis patients from the German calciphylaxis registry were compared with 370 dialysis patients without history of CUA. Genotyping was performed using iPLEX Gold MassARRAY(Sequenom, San Diego, USA), KASP genotyping chemistry (LGC, Teddington, Middlesex, UK) or sequencing. Statistical analysis comprised logistic regression analysis with adjustment for age and sex. Results: 165 SNPs were finally analyzed and 6 SNPs were associated with higher probability for calciphylaxis (OR>1) in our cohort. Nine SNPs of three genes (CD73, FGF23 and Vitamin D receptor) reached nominal significance (p< 0.05), but did not reach statistical significance after correction for multiple testing. Of the CD73 gene, rs4431401 (OR = 1.71, 95%CI 1.08-2.17, p = 0.023) and rs9444348 (OR = 1.48, 95% CI 1.11-1.97, p = 0.008) were associated with a higher probability for CUA. Of the FGF23 and VDR genes, rs7310492, rs11063118, rs13312747 and rs17882106 were associated with a higher probability for CUA. Conclusion: Polymorphisms in the genes encoding CD73, vitamin D receptor and FGF23 may play a role in calciphylaxis development. Although our study is the largest genetic study on calciphylaxis, it is limited by the low sample sizes. It therefore requires replication in other cohorts if available. 2017 PLoS One 12 2 urn:nbn:de:bvb:20-opus-171817 10.1371/journal.pone.0172407 Medizinische Klinik und Poliklinik I OPUS4-21119 Wissenschaftlicher Artikel Schmid, Rafael; Schmidt, Sonja K.; Hazur, Jonas; Detsch, Rainer; Maurer, Evelyn; Boccaccini, Aldo R.; Hauptstein, Julia; Teßmar, Jörg; Blunk, Torsten; Schrüfer, Stefan; Schubert, Dirk W.; Horch, Raymund E.; Bosserhoff, Anja K.; Arkudas, Andreas; Kengelbach-Weigand, Annika Comparison of hydrogels for the development of well-defined 3D cancer models of breast cancer and melanoma Bioprinting offers the opportunity to fabricate precise 3D tumor models to study tumor pathophysiology and progression. However, the choice of the bioink used is important. In this study, cell behavior was studied in three mechanically and biologically different hydrogels (alginate, alginate dialdehyde crosslinked with gelatin (ADA-GEL), and thiol-modified hyaluronan (HA-SH crosslinked with PEGDA)) with cells from breast cancer (MDA-MB-231 and MCF-7) and melanoma (Mel Im and MV3), by analyzing survival, growth, and the amount of metabolically active, living cells via WST-8 labeling. Material characteristics were analyzed by dynamic mechanical analysis. Cell lines revealed significantly increased cell numbers in low-percentage alginate and HA-SH from day 1 to 14, while only Mel Im also revealed an increase in ADA-GEL. MCF-7 showed a preference for 1% alginate. Melanoma cells tended to proliferate better in ADA-GEL and HA-SH than mammary carcinoma cells. In 1% alginate, breast cancer cells showed equally good proliferation compared to melanoma cell lines. A smaller area was colonized in high-percentage alginate-based hydrogels. Moreover, 3% alginate was the stiffest material, and 2.5% ADA-GEL was the softest material. The other hydrogels were in the same range in between. Therefore, cellular responses were not only stiffness-dependent. With 1% alginate and HA-SH, we identified matrices that enable proliferation of all tested tumor cell lines while maintaining expected tumor heterogeneity. By adapting hydrogels, differences could be accentuated. This opens up the possibility of understanding and analyzing tumor heterogeneity by biofabrication. 2020 Cancers 12 8 urn:nbn:de:bvb:20-opus-211195 10.3390/cancers12082320 Klinik und Poliklinik für Unfall-, Hand-, Plastische und Wiederherstellungschirurgie (Chirurgische Klinik II) OPUS4-11146 Dissertation Schmitt, Alexandra Role of Peroxiredoxin 6 in human melanoma Peroxiredoxin 6 (PRDX6) is a bifunctional enzyme comprising a peroxidase and a Ca2+-independent phospholipase (iPLA2) activity. This renders the enzyme capable of detoxifying reactive oxygen species (ROS) and of catalyzing the liberation of arachidonic acid (AA) from cellular membranes. Released AA can be further metabolized to bioactive lipids including eicosanoids, which are involved in inflammation, cell growth, differentiation, invasion and proliferation. Human melanoma cells are often characterized by imbalances in both ROS and lipid levels, which can be generated by oncogenic signaling, altered metabolism or UV irradiation. In previous studies, a comparative proteome analysis of the Xiphophorus fish melanoma model revealed a strong upregulation of Prdx6 in benign and malignant lesions compared to healthy skin. As the Xiphophorus melanoma model displays in many respects molecular characteristics that are similar to human melanoma, I investigated the functional role of PRDX6 in human melanoma cells. The first part of the study deals with the regulation of PRDX6 in melanocytes and human melanoma cells. I could demonstrate that the protein level of PRDX6 was strongly enhanced by the induction of the EGFR orthologue Xmrk from the Xiphophorus fish as well as the human EGFR. The upregulation of PRDX6 was further shown to be mediated in a PI3K-dependent and ROS-independent manner. The main part of the thesis comprises the investigation of the functional role of PRDX6 in human melanoma cells as well as the analysis of the underlying mechanism. I could show that knockdown of PRDX6 enhanced the oxidative stress response and led to decreased proliferation of melanoma cells. This cell growth effect was mainly mediated by the iPLA2 activity of PRDX6. Under conditions of strongly enhanced oxidative stress, the peroxidase activity became also important for cellular proliferation. Furthermore, the anti-proliferative effect in cells with lowered PRDX6 levels was the result of reduced cellular AA content and the decrease in the activation of SRC family proteins. Similarly, supplementation with AA led to regeneration of SRC family kinase activity and to an improvement in the reduced proliferation after knockdown of PRDX6. Since AA can be further processed into the prostaglandin PGE2, which has a pro-tumorigenic function in some cancer types, I further examined whether this eicosanoid is involved in the proliferative function of PRDX6. In contrast to AA, PGE2 was not consistently required for melanoma proliferation. In summary, I could demonstrate that PRDX6 plays a major role in AA-dependent lipid signaling in melanoma cells and thereby regulates proliferation. Interestingly, the proliferation relevant iPLA2 activity can be pharmacologically targeted, and melanoma cell growth was clearly blocked by the inhibitor BEL. Thus, I could identify the phospholipase activity of PRDX6 as a new therapeutically interesting target for melanoma treatment. 2015 urn:nbn:de:bvb:20-opus-111465 Theodor-Boveri-Institut für Biowissenschaften OPUS4-12060 Wissenschaftlicher Artikel Schrama, David; Ugurel, Selma; Sucker, Antje; Ritter, Cathrin; Zapatka, Marc; Schadendorf, Dirk; Becker, Jürgen Christian STAT3 Single Nucleotide Polymorphism rs4796793 SNP Does Not Correlate with Response to Adjuvant IFNα Therapy in Stage III Melanoma Patients Interferon alpha (IFNα) is approved for adjuvant treatment of stage III melanoma in Europe and the US. Its clinical efficacy, however, is restricted to a subpopulation of patients while side effects occur in most of treated patients. Thus, the identification of predictive biomarkers would be highly beneficial to improve the benefit to risk ratio. In this regard, STAT3 is important for signaling of the IFNα receptor. Moreover, the STAT3 single-nucleotide polymorphism (SNP) rs4796793 has recently been reported to be associated with IFNα sensitivity in metastatic renal cell carcinoma. To translate this notion to melanoma, we scrutinized the impact of rs4796793 functionally and clinically in this cancer. Interestingly, melanoma cells carrying the minor allele of rs4796793 were the most sensitive to IFNα in vitro. However, we did not detect a correlation between SNP genotype and STAT3 mRNA expression for either melanoma cells or for peripheral blood lymphocytes. Next, we analyzed the impact of rs4796793 on the clinical outcome of 259 stage III melanoma patients of which one-third had received adjuvant IFNα treatment. These analyses did not reveal a significant association between the STAT3 rs4796793 SNP and patients' progression free or overall survival when IFNα treated and untreated patients were compared. In conclusion, STAT3 rs4796793 SNP is no predictive marker for the efficacy of adjuvant IFNα treatment in melanoma patients. 2014 Frontiers in Medicine 1 47 urn:nbn:de:bvb:20-opus-120602 10.3389/fmed.2014.00047 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-31840 Wissenschaftlicher Artikel Schummer, Patrick; Schilling, Bastian How representative are data from global trials on programmed death-1 blockade in melanoma? 2022 1 The British Journal of Dermatology 187 3 283 284 urn:nbn:de:bvb:20-opus-318406 10.1111/bjd.21621 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-12683 Wissenschaftlicher Artikel Tomei, Sara; Adams, Sharon; Uccellini, Lorenzo; Bedognetti, Davide; De Giorgi, Valeria; Erdenebileg, Narnygerel; Libera Ascierto, Maria; Reinboth, Jennifer; Liu, Qiuzhen; Bevilacqua, Generoso; Wang, Ena; Mazzanti, Chiara; Marincola, Francesco M. Association between HRAS rs12628 and rs112587690 polymorphisms with the risk of melanoma in the North American population HRAS belongs to the RAS genes superfamily. RAS genes are important players in several human tumors and the single-nucleotide polymorphism rs12628 has been shown to contribute to the risk of bladder, colon, gastrointestinal, oral, and thyroid carcinoma. We hypothesized that this SNP may affect the risk of cutaneous melanoma as well. HRAS gene contains a polymorphic region (rs112587690), a repeated hexanucleotide -GGGCCT- located in intron 1. Three alleles of this region, P1, P2, and P3, have been identified that contain two, three, and four repeats of the hexanucleotide, respectively. We investigated the clinical impact of these polymorphisms in a case-control study. A total of 141 melanoma patients and 118 healthy donors from the North America Caucasian population were screened for rs12628 and rs112587690 polymorphisms. Genotypes were assessed by capillary sequencing or fragment analysis, respectively, and rs12628 CC and rs112587690 P1P1 genotypes significantly associated with increased melanoma risk (OR = 3.83, p = 0.003; OR = 11.3, p = 0.033, respectively), while rs112587690 P1P3 frequency resulted significantly higher in the control group (OR = 0.5, p = 0.017). These results suggest that rs12628 C homozygosis may be considered a potential risk factor for melanoma development in the North American population possibly through the linkage to rs112587690. 2012 3456-3461 Medical Oncology 29 5 urn:nbn:de:bvb:20-opus-126834 dx.doi.org/10.1007/s12032-012-0255-3 Theodor-Boveri-Institut für Biowissenschaften OPUS4-27513 Wissenschaftlicher Artikel Wendlinger, Simone; Wohlfarth, Jonas; Kreft, Sophia; Siedel, Claudia; Kilian, Teresa; Dischinger, Ulrich; Heppt, Markus V.; Wistuba-Hamprecht, Kilian; Meier, Friedegund; Goebeler, Matthias; Schadendorf, Dirk; Gesierich, Anja; Kosnopfel, Corinna; Schilling, Bastian Blood eosinophils are associated with efficacy of targeted therapy in patients with advanced melanoma Background: Eosinophils appear to contribute to the efficacy of immunotherapy and their frequency was suggested as a predictive biomarker. Whether this observation could be transferred to patients treated with targeted therapy remains unknown. Methods: Blood and serum samples of healthy controls and 216 patients with advanced melanoma were prospectively and retrospectively collected. Freshly isolated eosinophils were phenotypically characterized by flow cytometry and co-cultured in vitro with melanoma cells to assess cytotoxicity. Soluble serum markers and peripheral blood counts were used for correlative studies. Results: Eosinophil-mediated cytotoxicity towards melanoma cells, as well as phenotypic characteristics, were similar when comparing healthy donors and patients. However, high relative pre-treatment eosinophil counts were significantly associated with response to MAPKi (p = 0.013). Eosinophil-mediated cytotoxicity towards melanoma cells is dose-dependent and requires proximity of eosinophils and their target in vitro. Treatment with targeted therapy in the presence of eosinophils results in an additive tumoricidal effect. Additionally, melanoma cells affected eosinophil phenotype upon co-culture. Conclusion: High pre-treatment eosinophil counts in advanced melanoma patients were associated with a significantly improved response to MAPKi. Functionally, eosinophils show potent cytotoxicity towards melanoma cells, which can be reinforced by MAPKi. Further studies are needed to unravel the molecular mechanisms of our observations. 2022 Cancers 14 9 urn:nbn:de:bvb:20-opus-275137 10.3390/cancers14092294 Klinik und Poliklinik für Dermatologie, Venerologie und Allergologie OPUS4-5364 Wissenschaftlicher Artikel Wittbrodt, Joachim; Lammers, Reiner; Malitschek, Barbara; Ullrich, Axel; Schartl, Manfred Xmrk receptor tyrosine kinase is activated in Xiphophorus malignant melanoma Xmrk encodes a putative transmembrane glycoprotein of the tyrosine kinase family and is a melanoma-inducing gene in Xiphophorus. We attempted to investigate the biological function of the putative Xmrk receptor by characterizing its signalling properties. Since a potential Iigand for Xmrk has not yet been identified, it has been difficult to analyse the biochemical properlies and biological function of this cell surface protein. In an approach towards such analyses, the Xmrk extracellular domain was replaced by the closely related Iigand-binding domain sequences of the human epidennal growth factor receptor (HER) and the ligand-induced activity of the chimeric HER-Xmrk proteinwas examined. We show that the Xmrk protein is a functional receptor tyrosine kinase, is highly active in malignant melanoma and displays a constitutive autophosphorylation activity possibly due to an activating mutation in its extracellular or transmembrane domain. In the focus formation assay the HER-Xmrk chimera is a potent transfonning protein equivalent to other tyrosine kinase oncoproteins. 1992 urn:nbn:de:bvb:20-opus-61699 Theodor-Boveri-Institut für Biowissenschaften