@phdthesis{Alexander2019, author = {Alexander, Stephanie}, title = {Collective cancer cell invasion \(in\) \(vivo\): function of β1 and β3 integrins in perivascular invasion and resistance to therapy}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-85435}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2019}, abstract = {Pro-migratory signals mediated by the tumor microenvironment contribute to the cancer progression cascade, including invasion, metastasis and resistance to therapy. Derived from in vitro studies, isolated molecular steps of cancer invasion programs have been identified but their integration into the tumor microenvironment and suitability as molecular targets remain elusive. The purpose of the study was to visualize central aspects of tumor progression, including proliferation, survival and invasion by real-time intravital microscopy. The specific aims were to monitor the kinetics, mode, adhesion and chemoattraction mechanisms of tumor cell invasion, the involved guidance structures, and the response of invasion zones to anti-cancer therapy. To reach deeper tumor regions by optical imaging with subcellular resolution, near-infrared and infrared excited multiphoton microscopy was combined with a modified dorsal skinfold chamber model. Implanted HT-1080 fibrosarcoma and B16/F10 and MV3 melanoma tumors developed zones of invasive growth consisting of collective invasion strands that retained cell-cell contacts and high mitotic activity while invading at velocities of up to 200 μm per day. Collective invasion occurred predominantly along preexisting tissue structures, including blood and lymph vessels, collagen fibers and muscle strands of the deep dermis, and was thereby insensitive to RNAi based knockdown and/or antibody-based treatment against β1 and β3 integrins, chemokine (SDF-1/CXCL12) and growth factor (EGF) signaling. Therapeutic hypofractionated irradiation induced partial to complete regression of the tumor main mass, yet failed to eradicate the collective invasion strands, suggesting a microenvironmentally privileged niche. Whereas no radiosensitization was achieved by interference with EGFR or doxorubicin, the simultaneous inhibition of β1 and β3 integrins impaired cell proliferation and survival in spontaneously growing tumors and strongly enhanced the radiation response up to complete eradication of both main tumor and invasion strands. In conclusion, collective invasion in vivo is a robust process which follows preexisting tissue structures and is mainly independent of established adhesion and chemoattractant signaling. Due to its altered biological response to irradiation, collective invasion strands represent a microenvironmentally controlled and clinically relevant resistance niche to therapy. Therefore supportive regimens, such as anoikisinduction by anti-integrin therapy, may serve to enhance radio- and chemoefficacy and complement classical treatment regimens.}, subject = {Tumorzelle}, language = {en} } @article{KolbMaeurerSunderkoetterKukowskietal.2019, author = {Kolb-M{\"a}urer, Annette and Sunderk{\"o}tter, Cord and Kukowski, Borries and Meuth, Sven G.}, title = {An update on Peginterferon beta-1a Management in Multiple Sclerosis: results from an interdisciplinary Board of German and Austrian Neurologists and dermatologists}, series = {BMC Neurology}, volume = {19}, journal = {BMC Neurology}, doi = {10.1186/s12883-019-1354-y}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-224646}, year = {2019}, abstract = {Background: Interferon (IFN) beta drugs have been approved for the treatment of relapsing forms of multiple sclerosis (RMS) for more than 20years and are considered to offer a favourable benefit-risk profile. In July 2014, subcutaneous (SC) peginterferon beta-1a 125g dosed every 2weeks, a pegylated form of interferon beta-1a, was approved by the EMA for the treatment of adult patients with RRMS and in August 2014 by the FDA for RMS. Peginterferon beta-1a shows a prolonged half-life and increased systemic drug exposure resulting in a reduced dosing frequency compared to other available interferon-based products in MS. In the Phase 3 ADVANCE trial peginterferon beta-1a demonstrated significant positive effects on clinical and MRI outcome measures versus placebo after one year. Furthermore, in the ATTAIN extension study, sustained efficacy with long-term treatment for nearly 6years was shown. Main text In July 2016, an interdisciplinary panel of German and Austrian experts convened to discuss the management of side effects associated with peginterferon beta-1a and other interferon beta-based treatments in MS in daily practice. The panel was composed of experts from university hospitals and private clinics comprised of neurologists, dermatologists, and an MS nurse. In this paper we report recommendations regarding best practices for adverse event management, focussing on peginterferon beta-1a. Injection site reactions (ISRs) and influenza-like illness are the most common adverse effects of interferon beta therapies and can present a burden for MS patients leading to non-adherence and discontinuation of therapy. Peginterferon beta-1a shows improved pharmacological properties. In clinical trials, the adverse event (AE) profile of peginterferon beta-1a was similar to other interferon beta formulations. The most common AEs were mild to moderate ISRs, influenza-like illness, pyrexia, and headache. Current information on the underlying cause of skin reactions associated with SC interferon treatment, and the management strategies for these AEs are limited. In pivotal trials, ISRs were mainly characterized and classified by neurologists, while dermatologists were only rarely consulted. Conclusions This report addresses expert recommendations on the management of most relevant adverse effects related to peginterferon beta-1a and other interferon betas, based on literature and interdisciplinary experience.}, language = {en} } @article{RauschenbergerSchmittAzeemetal.2019, author = {Rauschenberger, Tabea and Schmitt, Viola and Azeem, Muhammad and Klein-Hessling, Stefan and Murti, Krisna and Gr{\"a}n, Franziska and Goebeler, Matthias and Kerstan, Andreas and Klein, Matthias and Bopp, Tobias and Serfling, Edgar and Muhammad, Khalid}, title = {T cells control chemokine secretion by keratinocytes}, series = {Frontiers in Immunology}, volume = {10}, journal = {Frontiers in Immunology}, number = {1917}, issn = {1664-3224}, doi = {10.3389/fimmu.2019.01917}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-195695}, year = {2019}, abstract = {The massive infiltration of lymphocytes into the skin is a hallmark of numerous human skin disorders. By co-culturing murine keratinocytes with splenic T cells we demonstrate here that T cells affect and control the synthesis and secretion of chemokines by keratinocytes. While pre-activated CD8\(^+\)T cells induce the synthesis of CXCL9 and CXCL10 in keratinocytes and keep in check the synthesis of CXCL1, CXCL5, and CCL20, keratinocytes dampen the synthesis of CCL3 and CCL4 in pre-activated CD8\(^+\)T cells. One key molecule is IFN-γ that is synthesized by CD8\(^+\)T cells under the control of NFATc1 and NFATc2. CD8\(^+\)T cells deficient for both NFAT factors are unable to induce CXCL9 and CXCL10 expression. In addition, CD8\(^+\)T cells induced numerous type I IFN-inducible "defense genes" in keratinocytes encoding the PD1 and CD40 ligands, TNF-α and caspase-1. The enhanced expression of type I IFN-inducible genes resembles the gene expression pattern at the dermal/epidermal interface in lichen planus, an inflammatory T lymphocyte-driven skin disease, in which we detected the expression of CXCL10 in keratinocytes in close vicinity to the infiltration front of T cells. These data reflect the multifaceted interplay of lymphocytes with keratinocytes at the molecular level.}, language = {en} } @article{KrebsBehrmannKalogirouetal.2019, author = {Krebs, Markus and Behrmann, Christoph and Kalogirou, Charis and Sokolakis, Ioannis and Kneitz, Susanne and Kruithof-de Julio, Marianna and Zoni, Eugenio and Rech, Anne and Schilling, Bastian and K{\"u}bler, Hubert and Spahn, Martin and Kneitz, Burkhard}, title = {miR-221 Augments TRAIL-mediated apoptosis in prostate cancer cells by inducing endogenous TRAIL expression and targeting the functional repressors SOCS3 and PIK3R1}, series = {BioMed Research International}, volume = {2019}, journal = {BioMed Research International}, doi = {10.1155/2019/6392748}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-202480}, pages = {6392748}, year = {2019}, abstract = {miR-221 is regarded as an oncogene in many malignancies, and miR-221-mediated resistance towards TRAIL was one of the first oncogenic roles shown for this small noncoding RNA. In contrast, miR-221 is downregulated in prostate cancer (PCa), thereby implying a tumour suppressive function. By using proliferation and apoptosis assays, we show a novel feature of miR-221 in PCa cells: instead of inducing TRAIL resistance, miR-221 sensitized cells towards TRAIL-induced proliferation inhibition and apoptosis induction. Partially responsible for this effect was the interferon-mediated gene signature, which among other things contained an endogenous overexpression of the TRAIL encoding gene TNFSF10. This TRAIL-friendly environment was provoked by downregulation of the established miR-221 target gene SOCS3. Moreover, we introduced PIK3R1 as a target gene of miR-221 in PCa cells. Proliferation assays showed that siRNA-mediated downregulation of SOCS3 and PIK3R1 mimicked the effect of miR-221 on TRAIL sensitivity. Finally, Western blotting experiments confirmed lower amounts of phospho-Akt after siRNA-mediated downregulation of PIK3R1 in PC3 cells. Our results further support the tumour suppressing role of miR-221 in PCa, since it sensitises PCa cells towards TRAIL by regulating the expression of the oncogenes SOCS3 and PIK3R1. Given the TRAIL-inhibiting effect of miR-221 in various cancer entities, our results suggest that the influence of miR-221 on TRAIL-mediated apoptosis is highly context- and entity-dependent.}, language = {en} } @article{BallinHotzBourratetal.2019, author = {Ballin, Nadja and Hotz, Alrun and Bourrat, Emmanuelle and K{\"u}sel, Julia and Oji, Vinzenz and Bouadjar, Bakar and Brognoli, Davide and Hickman, Geoffroy and Heinz, Lisa and Vabres, Pierre and Marrakchi, Slaheddine and Leclerc-Mercier, St{\´e}phanie and Irvine, Alan and Tadini, Gianluca and Hamm, Henning and Has, Cristina and Blume-Peytavi, Ulrike and Mitter, Diana and Reitenbach, Marina and Hausser, Ingrid and Zimmer, Andreas D. and Alter, Svenja and Fischer, Judith}, title = {Genetical, clinical, and functional analysis of a large international cohort of patients with autosomal recessive congenital ichthyosis due to mutations in NIPAL4}, series = {Human Mutation}, volume = {40}, journal = {Human Mutation}, number = {12}, doi = {10.1002/humu.23883}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-212747}, pages = {2318-2333}, year = {2019}, abstract = {Autosomal recessive congenital ichthyosis (ARCI) belongs to a heterogeneous group of disorders of keratinization. To date, 10 genes have been identified to be causative for ARCI. NIPAL4 (Nipa-Like Domain-Containing 4) is the second most commonly mutated gene in ARCI. In this study, we present a large cohort of 101 families affected with ARCI carrying mutations in NIPAL4. We identified 16 novel mutations and increase the total number of pathogenic mutations in NIPAL4 to 34. Ultrastructural analysis of biopsies from six patients showed morphological abnormalities consistent with an ARCI EM type III. One patient with a homozygous splice site mutation, which leads to a loss of NIPAL4 mRNA, showed additional ultrastructural aberrations together with a more severe clinical phenotype. Our study gives insights into the frequency of mutations, a potential hot spot for mutations, and genotype-phenotype correlations.}, language = {en} } @article{ThiemHesbacherKneitzetal.2019, author = {Thiem, Alexander and Hesbacher, Sonja and Kneitz, Hermann and di Primio, Teresa and Heppt, Markus V. and Hermanns, Heike M. and Goebeler, Matthias and Meierjohann, Svenja and Houben, Roland and Schrama, David}, title = {IFN-gamma-induced PD-L1 expression in melanoma depends on p53 expression}, series = {Journal of Experimental \& Clinical Cancer Research}, volume = {38}, journal = {Journal of Experimental \& Clinical Cancer Research}, doi = {10.1186/s13046-019-1403-9}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-201016}, pages = {397}, year = {2019}, abstract = {Background Immune checkpoint inhibition and in particular anti-PD-1 immunotherapy have revolutionized the treatment of advanced melanoma. In this regard, higher tumoral PD-L1 protein (gene name: CD274) expression is associated with better clinical response and increased survival to anti-PD-1 therapy. Moreover, there is increasing evidence that tumor suppressor proteins are involved in immune regulation and are capable of modulating the expression of immune checkpoint proteins. Here, we determined the role of p53 protein (gene name: TP53) in the regulation of PD-L1 expression in melanoma. Methods We analyzed publicly available mRNA and protein expression data from the cancer genome/proteome atlas and performed immunohistochemistry on tumors with known TP53 status. Constitutive and IFN-ɣ-induced PD-L1 expression upon p53 knockdown in wildtype, TP53-mutated or JAK2-overexpressing melanoma cells or in cells, in which p53 was rendered transcriptionally inactive by CRISPR/Cas9, was determined by immunoblot or flow cytometry. Similarly, PD-L1 expression was investigated after overexpression of a transcriptionally-impaired p53 (L22Q, W23S) in TP53-wt or a TP53-knockout melanoma cell line. Immunoblot was applied to analyze the IFN-ɣ signaling pathway. Results For TP53-mutated tumors, an increased CD274 mRNA expression and a higher frequency of PD-L1 positivity was observed. Interestingly, positive correlations of IFNG mRNA and PD-L1 protein in both TP53-wt and -mutated samples and of p53 and PD-L1 protein suggest a non-transcriptional mode of action of p53. Indeed, cell line experiments revealed a diminished IFN-ɣ-induced PD-L1 expression upon p53 knockdown in both wildtype and TP53-mutated melanoma cells, which was not the case when p53 wildtype protein was rendered transcriptionally inactive or by ectopic expression of p53\(^{L22Q,W23S}\), a transcriptionally-impaired variant, in TP53-wt cells. Accordingly, expression of p53\(^{L22Q,W23S}\) in a TP53-knockout melanoma cell line boosted IFN-ɣ-induced PD-L1 expression. The impaired PD-L1-inducibility after p53 knockdown was associated with a reduced JAK2 expression in the cells and was almost abrogated by JAK2 overexpression. Conclusions While having only a small impact on basal PD-L1 expression, both wildtype and mutated p53 play an important positive role for IFN-ɣ-induced PD-L1 expression in melanoma cells by supporting JAK2 expression. Future studies should address, whether p53 expression levels might influence response to anti-PD-1 immunotherapy.}, language = {en} } @phdthesis{Geiseler2019, author = {Geiseler, Marina}, title = {Klinische, histopathologische und immunph{\"a}notypische Charakterisierung der CD4+ klein-/mittelgroßzelligen kutanen T-Zell-Lymphoproliferation}, doi = {10.25972/OPUS-18411}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-184112}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2019}, abstract = {Innerhalb der Gruppe der kutanen T-Zell-Lymphome ist die CD4+ klein-/mittelgroßzellige T-Zell-Lymphoproliferation (SMTCL) eine seltene und bisher als provisorisch erfasste Entit{\"a}t. Um genauere Kenntnisse zu Klinik und Verlauf, Histologie und Immunph{\"a}notyp zu gewinnen, untersuchten wir in dieser Fallserie an 95 F{\"a}llen entsprechende Charakteristika. Dabei konnten alle der bisher provisorisch definierten Eigenschaften der SMTCL best{\"a}tigt werden. Lediglich die 5-Jahre-{\"U}berlebensrate zeigte sich mit 100\% weitaus h{\"o}her als die in der WHO-/EORTC-Klassifikation angegebenen 60-80\%. Diese Best{\"a}tigung der provisorisch definierten Eigenschaften an einem gr{\"o}ßeren Kollektiv kann dazu beitragen, dass die SMTCL in die n{\"a}chste Version der WHO-Klassifikation - durch ausreichende Daten gest{\"u}tzt - als definitive Entit{\"a}t aufgenommen werden kann. Insgesamt konnten in dieser Studie einige Faktoren, die mit einem weniger indolenten Krankheitsverlauf assoziiert sind, identifiziert werden. So zeigten klinisch ein initial bestehender generalisierter Hautbefall sowie eine extrafaziale Lokalisation der L{\"a}sion einen weniger indolenten Verlauf an. Auch Patienten, bei denen ein Verlust von CD2, CD3 oder CD5 bei den Tumorzellen festgestellt wurde, wiesen einen f{\"u}r die CD4+ SMTCL ungew{\"o}hnlichen und weniger indolenten Krankheitsverlauf mit h{\"a}ufigeren Rezidiven und seltenerem Erreichen einer kompletten Remission auf. Histopathologisch schien eine oberfl{\"a}chlichere Infiltrattiefe des Pr{\"a}parates sowie das Vorhandensein eines fokalen Epidermotropismus einen negativen prognostischen Wert zu besitzen. Bez{\"u}glich des Gesamt{\"u}berlebens hatten allerdings auch Patienten mit einem der identifizierten negativen prognostischen Faktoren eine exzellente Prognose. Bei Vorliegen eines dieser negativen Faktoren sollte jedoch eine engmaschigere klinische {\"U}berwachung erfolgen.}, subject = {Hautlymphom}, language = {de} } @article{NothhaftKlepperKneitzetal.2019, author = {Nothhaft, Matthias and Klepper, Joerg and Kneitz, Hermann and Meyer, Thomas and Hamm, Henning and Morbach, Henner}, title = {Hemorrhagic bullous Henoch-Sch{\"o}nlein Purpura: case report and review of the literature}, series = {Frontiers in Pediatrics}, volume = {6}, journal = {Frontiers in Pediatrics}, doi = {10.3389/fped.2018.00413}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-201435}, pages = {413}, year = {2019}, abstract = {Henoch-Sch{\"o}nlein Purpura (HSP) or IgA vasculitis is the most common systemic vasculitis of childhood and may affect skin, joints, gastrointestinal tract, and kidneys. Skin manifestations of HSP are characteristic and include a non-thrombocytopenic palpable purpura of the lower extremities and buttocks. Rarely, HSP may initially present as or evolve into hemorrhagic vesicles and bullae. We present an otherwise healthy 5-year-old boy with an acute papulovesicular rash of both legs and intermittent abdominal pain. After a few days the skin lesions rapidly evolved into palpable purpura and hemorrhagic bullous lesions of variable size and severe hemorrhagic HSP was suspected. A histological examination of a skin biopsy showed signs of a small vessel leukocytoclastic vasculitis limited to the upper dermis and direct immunofluorescence analysis revealed IgA deposits in vessel walls, compatible with HSP. To further characterize the clinical picture and treatment options of bullous HSP we performed an extensive literature research and identified 41 additional pediatric patients with bullous HSP. Two thirds of the reported patients were treated with systemic corticosteroids, however, up to 25\% of the reported patients developed skin sequelae such as hyperpigmentation and/or scarring. The early use of systemic corticosteroids has been discussed controversially and suggested in some case series to be beneficial by reducing the extent of lesions and minimizing sequelae of disease. Our patient was treated with systemic corticosteroids tapered over 5 weeks. Fading of inflammation resulted in healing of most erosions, however, a deep necrosis developing from a large blister at the dorsum of the right foot persisted so that autologous skin transplantation was performed. Re-examination 11 months after disease onset showed complete clinical remission with re-epithelialization but also scarring of some affected areas.}, language = {en} } @article{WobserWeberGlunzetal.2019, author = {Wobser, Marion and Weber, Alexandra and Glunz, Amelie and Tauch, Saskia and Seitz, Kristina and Butelmann, Tobias and Hesbacher, Sonja and Goebeler, Matthias and Bartz, Ren{\´e} and Kohlhof, Hella and Schrama, David and Houben, Roland}, title = {Elucidating the mechanism of action of domatinostat (4SC-202) in cutaneous T cell lymphoma cells}, series = {Journal of Hematology \& Oncology}, volume = {12}, journal = {Journal of Hematology \& Oncology}, doi = {10.1186/s13045-019-0719-4}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-200703}, pages = {30}, year = {2019}, abstract = {Background Targeting epigenetic modifiers is effective in cutaneous T cell lymphoma (CTCL). However, there is a need for further improvement of this therapeutic approach. Here, we compared the mode of action of romidepsin (FK228), an established class I histone deacetylase inhibitor, and domatinostat (4SC-202), a novel inhibitor of class I HDACs, which has been reported to also target the lysine-specific histone demethylase 1A (LSD1). Methods We performed MTS assays and flow cytometric analyses of propidium iodide or annexin V-stained cells to assess drug impact on cellular proliferation, cell cycle distribution, and survival. Histone acetylation and methylation as well as caspase activation was analyzed by immunoblot. Gene expression analysis was performed using NanosString technology. Knockdown and knockout of LSD1 was achieved with shRNA and CRISPR/Cas9, respectively, while the CRISPR/Cas9 synergistic activation mediator system was used to induce expression of endogenous HDACs and LSD1. Furthermore, time-lapse fluorescence microscopy and an in vitro tubulin polymerization assay were applied. Results While FK228 as well as 4SC-202 potently induced cell death in six different CTCL cell lines, only in the case of 4SC-202 death was preceded by an accumulation of cells in the G2/M phase of the cell cycle. Surprisingly, apoptosis and accumulation of cells with double DNA content occurred already at 4SC-202 concentrations hardly affecting histone acetylation and methylation, and provoking significantly less changes in gene expression compared to biologically equivalent doses of FK228. Indeed, we provide evidence that the 4SC-202-induced G2/M arrest in CTCL cells is independent of de novo transcription. Furthermore, neither enforced expression of HDAC1 nor knockdown or knockout of LSD1 affected the 4SC-202-induced effects. Since time-lapse microscopy revealed that 4SC-202 could affect mitotic spindle formation, we performed an in vitro tubulin polymerization assay revealing that 4SC-202 can directly inhibit microtubule formation. Conclusions We demonstrate that 4SC-202, a drug currently tested in clinical trials, effectively inhibits growth of CTCL cells. The anti-cancer cell activity of 4SC-202 is however not limited to LSD1-inhibition, modulation of histone modifications, and consecutive alteration of gene expression. Indeed, the compound is also a potent microtubule-destabilizing agent.}, language = {en} } @article{GlutschGraenWeberetal.2019, author = {Glutsch, Valerie and Gr{\"a}n, Franziska and Weber, Judith and Gesierich, Anja and Goebeler, Matthias and Schilling, Bastian}, title = {Response to combined ipilimumab and nivolumab after development of a nephrotic syndrome related to PD-1 monotherapy}, series = {Journal for ImmunoTherapy of Cancer}, volume = {7}, journal = {Journal for ImmunoTherapy of Cancer}, doi = {10.1186/s40425-019-0655-4}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-201214}, pages = {181}, year = {2019}, abstract = {Background High response rates of metastatic melanoma have been reported upon immune checkpoint inhibition by PD-1 blockade alone or in combination with CTLA-4 inhibitors. However, the majority of patients with a primary resistance to anti-PD-1 monotherapy is also refractory to a subsequent combined checkpoint inhibition. In BRAF wildtype patients with a primary resistance to PD-1 inhibitors, therapeutic options are therefore limited and immune-related adverse events (irAE) have to be taken into consideration when discussing a subsequent immunotherapy. Case presentation We report the case of a 68-year-old male patient with metastatic melanoma who experienced an acute renal failure with nephrotic syndrome due to a minimal change disease developing after a single dose of the anti-PD-1 antibody pembrolizumab. A kidney biopsy revealed a podocytopathy without signs of interstitial nephritis. Renal function recovered to almost normal creatinine and total urine protein levels upon treatment with oral steroids and diuretics. Unfortunately, a disease progression (PD, RECIST 1.1) was observed in a CT scan after resolution of the irAE. In a grand round, re-exposure to a PD-1-containing regime was recommended. Consensually, a combined immunotherapy with ipilimumab and nivolumab was initiated. Nephrotoxicity was tolerable during combined immunotherapy and a CT scan of chest and abdomen showed a deep partial remission (RECIST 1.1) after three doses of ipilimumab (3 mg/kg) and nivolumab (1 mg/kg). Conclusion This case illustrates that a fulminant response to combined checkpoint inhibition is possible after progression after anti-PD-1 monotherapy and a severe irAE.}, language = {en} }