@phdthesis{Landwehr2023, author = {Landwehr, Laura-Sophie}, title = {Steroid Hormones and Cancer Immunity - learning from Adrenocortical Carcinoma}, doi = {10.25972/OPUS-25189}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-251895}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Adrenocortical carcinoma (ACC) is a rare, but highly aggressive endocrine malignancy. Tumor-related hypercortisolism is present in 60 \% of patients and associated with worse outcome. While cancer immunotherapies have revolutionized the treatment of many cancer entities, the results of initial studies of different immune checkpoint inhibitors in ACC were heterogeneous. Up to now, five small clinical trials with a total of 121 patients have been published and demonstrated an objective response in only 17 patients. However, one of the studies, by Raj et al., reported a clinically meaningful disease control rate of 52 \% and a median overall survival of almost 25 months suggesting that a subgroup of ACC patients may benefit from immunotherapeutic approaches. Following the hypothesis that some ACCs are characterized by a glucocorticoid-induced T lymphocytes depletion, several studies were performed as part of the presented thesis. First, the immune cell infiltration in a large cohort of 146 ACC specimens was investigated. It was demonstrated for the first time, and against the common assumption, that ACCs were infiltrated not only by FoxP3+ regulatory T cells (49.3 \%), but also that a vast majority of tumor samples was infiltrated by CD4+ TH cells (74 \%) and CD8+ cytotoxic T cells (84.3 \%), albeit the immune cell number varied heterogeneously and was rather low (median: 7.7 CD3+ T cells / high power field, range: 0.1-376). Moreover, the presence of CD3+-, CD4+- and CD8+ ACC-infiltrating lymphocytes was associated with an improved recurrence-free (HR: 0.31 95 \% CI 0.11-0.82) and overall survival (HR: 0.47 96 \% CI 0.25-0.87). Particularly, patients with tumor-infiltrating CD4+ TH cells without glucocorticoid excess had a significantly longer overall survival compared to patients with T cell-depleted ACC and hypercortisolism (121 vs. 27 months, p = 0.004). Hence, the impact of glucocorticoids might to some extent be responsible for the modest immunogenicity in ACC as hypercortisolism was reversely correlated with the number of CD4+ TH cells. Accordingly, CD3+ T cells co-cultured with steroidogenic NCI-H295R ACC cells demonstrated in vitro an enhanced anti-tumoral cytotoxicity by secreting 747.96 ±225.53 pg/ml IFN-γ in a therapeutically hormone-depleted microenvironment (by incubation with metyrapone), versus only 276.02 ±117.46 pg/ml IFN-γ in a standard environment with glucocorticoid excess. Other potential biomarkers to predict response to immunotherapies are the immunomodulatory checkpoint molecules, programmed cell death 1 (PD-1) and its ligand PD-L1, since both are targets of antibodies used therapeutically in different cancer entities. In a subcohort of 129 ACCs, expressions of both molecules were heterogeneous (PD-1 17.4 \%, range 1-15; PD-L1 24.4 \%, range 1 - 90) and rather low. Interestingly, PD-1 expression significantly influenced ACC patients´ overall (HR: 0.21 95 \% CI 0.53-0.84) and progression- free survival (HR: 0.30 95 \% CI 0.13-0.72) independently of established factors, like ENSAT tumor stage, resection status, Ki67 proliferation index and glucocorticoid excess, while PD-L1 had no impact. In conclusion, this study provides several potential explanations for the heterogeneous results of the immune checkpoint therapy in advanced ACC. In addition, the establishment of PD-1 as prognostic marker can be easily applied in routine clinical care, because it is nowadays anyway part of a detailed histo-pathological work-up. Furthermore, these results provide the rationale and will pave the way towards a combination therapy using immune checkpoint inhibitors as well as glucocorticoid blockers. This will increase the likelihood of re-activating the immunological anti-tumor potential in ACC. However, this will have to be demonstrated by additional preclinical in vivo experiments and finally in clinical trials with patients.}, subject = {Steroidhormon}, language = {en} } @phdthesis{Mestermann2020, author = {Mestermann, Katrin}, title = {Pharmacological control of CAR T-cells by dasatinib}, doi = {10.25972/OPUS-18056}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-180562}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2020}, abstract = {Cellular therapies using chimeric antigen receptor (CAR) modified T cells to eradicate tumor cells have been a major breakthrough in the treatment of hematologic malignancies. However, there are no measures to control CAR T cell activity after infusion, which is mostly required in cases of CAR T cell overreaction, e.g. cytokine release syndrome, or in the case of T cell failure, e.g. caused by exhaustion. In our study, we identified the tyrosine kinase inhibitor (TKI) dasatinib (© Sprycel) as a suitable agent to steer CAR T cells in vitro and in vivo. We show that single treatment of CD4+ and CD8+ CAR T cells with dasatinib conferred either partial or complete inhibition, depending on the applied concentration. The blockade was immediate and encompassed spe-cific lysis, cytokine secretion and proliferation following antigen encounter. The mechanism relied on reduced phosphorylation of key kinases in the CAR signaling cascade, which led to abrogation of nuclear factor of activated T-cells (NFAT) signaling. Importantly, inhibition was fully reversible by dasatinib withdrawal. In vivo, dasatinib blocked CAR T cell function without impairing the engraftment of CAR T cells or their subsequent anti tumor function once dasatinib administration was discontinued. We therefore introduce dasatinib as a new tool to efficiently block CAR T cells in vitro and in vivo, with data suggesting that dasatinib can be used in a clinical setting to mitigate toxicity after adaptive transfer of CAR modified T cells and other forms of T cell based immunotherapy. Additionally we show that intermittent inhibition of CAR T cells by dasatinib im-proves the efficacy of CAR T cell therapy. By pausing T cells for short periods of time in vi-vo, upregulation of programmed death protein 1 (PD-1) and subsequent induction of exhaus-tion was prevented, which increased the expansion of T cells and the rate of tumor eradica-tion. Our data therefore suggest that dasatinib can additionally be used to overcome T cell exhaustion that is induced by massive tumor burden and upregulation of inhibitory receptors.}, subject = {Immuntherapie}, language = {en} } @phdthesis{Marquardt2023, author = {Marquardt, Andr{\´e}}, title = {Machine-Learning-Based Identification of Tumor Entities, Tumor Subgroups, and Therapy Options}, doi = {10.25972/OPUS-32954}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-329548}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Molecular genetic analyses, such as mutation analyses, are becoming increasingly important in the tumor field, especially in the context of therapy stratification. The identification of the underlying tumor entity is crucial, but can sometimes be difficult, for example in the case of metastases or the so-called Cancer of Unknown Primary (CUP) syndrome. In recent years, methylome and transcriptome utilizing machine learning (ML) approaches have been developed to enable fast and reliable tumor and tumor subtype identification. However, so far only methylome analysis have become widely used in routine diagnostics. The present work addresses the utility of publicly available RNA-sequencing data to determine the underlying tumor entity, possible subgroups, and potential therapy options. Identification of these by ML - in particular random forest (RF) models - was the first task. The results with test accuracies of up to 99\% provided new, previously unknown insights into the trained models and the corresponding entity prediction. Reducing the input data to the top 100 mRNA transcripts resulted in a minimal loss of prediction quality and could potentially enable application in clinical or real-world settings. By introducing the ratios of these top 100 genes to each other as a new database for RF models, a novel method was developed enabling the use of trained RF models on data from other sources. Further analysis of the transcriptomic differences of metastatic samples by visual clustering showed that there were no differences specific for the site of metastasis. Similarly, no distinct clusters were detectable when investigating primary tumors and metastases of cutaneous skin melanoma (SKCM). Subsequently, more than half of the validation datasets had a prediction accuracy of at least 80\%, with many datasets even achieving a prediction accuracy of - or close to - 100\%. To investigate the applicability of the used methods for subgroup identification, the TCGA-KIPAN dataset, consisting of the three major kidney cancer subgroups, was used. The results revealed a new, previously unknown subgroup consisting of all histopathological groups with clinically relevant characteristics, such as significantly different survival. Based on significant differences in gene expression, potential therapeutic options of the identified subgroup could be proposed. Concludingly, in exploring the potential applicability of RNA-sequencing data as a basis for therapy prediction, it was shown that this type of data is suitable to predict entities as well as subgroups with high accuracy. Clinical relevance was also demonstrated for a novel subgroup in renal cell carcinoma. The reduction of the number of genes required for entity prediction to 100 genes, enables panel sequencing and thus demonstrates potential applicability in a real-life setting.}, subject = {Maschinelles Lernen}, language = {en} } @phdthesis{Hotz2008, author = {Hotz, Christian}, title = {Improvement of Salmonella vaccine strains for cancer immune therapy based on secretion or surface display of antigens}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-29548}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2008}, abstract = {Cancer immune therapy represents a promising alternative to conventional anti tumour therapy like radiation, surgical excision of the tumour or classical chemotherapy. The biggest advantage of cancer immune therapy is specificity, achieved by targeting tumour-associated antigens with the effector arms of the host immune system. This is believed to result in less adverse effects than standard therapy and reaches presumably also metastatic lesions at distant sites from the primary tumour. However, cancer immune therapy by vaccination against tumour antigens failed to translate into clinical success, yet. Furthermore, despite tremendous clinical efforts malignant disease still results in high mortalities giving rise to the need for novel vaccination-based therapies against cancer. An interesting approach in this respect is the use of bacteria like attenuated salmonellae as carriers for heterologous cancer antigens. In numerous preclinical studies Salmonella-based vaccines could elicit cell mediated immune responses of the CD4+ and CD8+ type against own and heterologous antigens which make them ideally suited for anti tumour therapy. Special delivery systems in Salmonella carriers like surface display or secretion of antigens were shown to be advantageous for the immunological outcome. This work focussed on developing novel Salmonella carriers for immune therapy against cancer. In a first project, TolC, a multifunctional outer membrane protein of E. coli was utilized as membrane anchor for 3 heterologous antigens. Respective TolC fusion proteins encoded on plasmids were analysed for expression, functionality and plasmid stability in different engineered Salmonella strains. The amount of membrane localized recombinant TolC was enhanced in tolC-deficient strains. Furthermore, fusion proteins were functional and plasmid stability was very high in vitro and in vivo. Disappointingly, neither specific CD4+/CD8+ T-cell responses against the model antigen ovalbumin nor CD8+ responses against the cancer antigen BRAFV600E were detectable in murine model systems. However, mice immunized with Salmonella strains displaying an immunodominant epitope of the cancer related prostate specific antigen (PSA) were partially protected from subsequent tumour challenge with a PSA expressing melanoma cell line. Tumour growth in mice immunized with the respective strain was significantly decelerated compared to controls, thus indicating that this surface display system confers protective immunity against tumours. In a second study, the approved typhoid vaccine strain Salmonella enterica serovar Typhi Ty21a (Ty21a) was improved for the hemolysin type I secretion system of E. coli. This secretion system is widely used for heterologous antigen delivery in live bacterial vaccines. It was demonstrated throughout this work that a mutation of rpoS in Ty21a correlated with decreased ability for hemolysin secretion compared to other Salmonella strains. Complementation with rpoS or the presumed downstream target of rpoS, rfaH resulted in enhanced expression and secretion of heterologous hemolysin in Ty21a. Presumably by raising the amount of free antigen, rfaHcomplemented Ty21a elicited higher antibody titres against heterologous hemolysin in immunized mice than controls and even rpoS-positive Ty21a. Therefore, rfaHcomplemented Ty21a could form the basis of a novel generation of vaccines for human use based on (cancer) antigen secretion.}, subject = {Impfstoff}, language = {en} } @phdthesis{Hein2014, author = {Hein, Melanie}, title = {Functional analysis of angiogenic factors in tumor cells and endothelia}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-93863}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2014}, abstract = {Tumor angiogenesis is essential for the growth of solid tumors as their proliferation and survival is dependent on consistent oxygen and nutrient supply. Anti-angiogenic treatments represent a therapeutic strategy to inhibit tumor growth by preventing the formation of new blood vessels leading to starvation of the tumor. One of the best characterized anti angiogenic therapeutics is the monoclonal antibody bevacizumab (Avastin), which targets and neutralizes VEGF leading to disruption of the VEGF signaling pathway. Until today, bevacizumab has found its way into clinical practice and has gained approval for treatment of different types of cancer including colorectal cancer, non-small cell lung cancer, breast cancer and renal cell carcinoma. Signaling of VEGF is mediated through VEGF receptors, mainly VEGFR2, which are primarily located on the cell surface of endothelial cells. However, there has been evidence that expression of VEGF receptors can also be found on tumor cells themselves raising the possibility of autocrine and/or paracrine signaling loops. Thus, tumor cells could also benefit from VEGF signaling, which would promote tumor growth. The aim of this study was to investigate if bevacizumab has a direct effect on tumor cells in vitro. To this end, tumor cell lines from the NCI-60 panel derived from four different tumor types were treated with bevacizumab and angiogenic gene and protein expression as well as biological outputs including proliferation, migration and apoptosis were investigated. Most of the experiments were performed under hypoxia to mimic the in vivo state of tumors. Overall, there was a limited measurable effect of bevacizumab on treated tumor cell lines according to gene and protein expression changes as well as biological functions when compared to endothelial controls. Minor changes in terms of proliferation or gene regulation were evident in a single tumor cell line after VEGF-A blockade by bevacizumab, which partially demonstrated a direct effect on tumor cells. However, the overall analysis revealed that tumor cell lines are not intrinsically affected in an adverse manner by bevacizumab treatment. Besides the functional analysis of tumor cells, embryonic stem cell derived endothelial cells were characterized to delineate vascular Hey gene functions. Hey and Hes proteins are the best characterized downstream effectors of the evolutionary conserved Notch signaling pathway, which mainly act as transcriptional repressors regulating downstream target genes. Hey proteins play a crucial role in embryonic development as loss of Hey1 and Hey2 in mice in vivo leads to a severe vascular phenotype resulting in early embryonic lethality. The major aim of this part of the thesis was to identify vascular Hey target genes using embryonic stem cell derived endothelial cells utilizing a directed endothelial differentiation approach, as ES cells and their differentiation ability provide a powerful in vitro system to study developmental processes. To this end, Hey deficient and Hey wildtype embryonic stem cells were stably transfected with an antibiotic selection marker driven by an endothelial specific promoter, which allows selection for endothelial cells. ESC-derived endothelial cells exhibited typical endothelial characteristics as shown by marker gene expression, immunofluorescent staining and tube formation ability. In a second step, Hey deficient ES cells were stably transfected with doxycycline inducible Flag-tagged Hey1 and Hey2 transgenes to re-express Hey proteins in the respective cell line. RNA-Sequencing of Hey deficient and Hey overexpressing ES cells as well as ESC-derived endothelial cells revealed many Hey downstream target genes in ES cells and fewer target genes in endothelial cells. Hey1 and Hey2 more or less redundantly regulate target genes in ES cells, but some genes were regulated by Hey2 alone. According to Gene Ontology term analysis, Hey target genes are mainly involved in embryonic development and transcriptional regulation. However, the response of ESC-derived endothelial cells in regulating Hey downstream target genes was rather limited when compared to ES cells, which could be due to lower transgene expression in endothelial cells. The limited response also raises the possibility that target gene regulation in endothelial cells is not only dependent on Hey gene functions alone and thus loss or overexpression of Hey genes in this in vitro setting does not influence target gene regulation.}, subject = {Krebs }, language = {en} } @phdthesis{Monjezi2018, author = {Monjezi, Razieh}, title = {Engineering of chimeric antigen receptor T cells with enhanced therapeutic index in cancer immunotherapy using non-viral gene transfer and genome editing}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-152521}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2018}, abstract = {The advances in genetic engineering have enabled us to confer T cells new desired functions or delete their specific undesired endogenous properties for improving their antitumor function. Due to their efficient gene delivery, viral vectors have been successfully used in T-cell engineering to provide gene transfer medicinal products for the treatment of human disease. One example is adoptive cell therapy with T cells that were genetically modified with gamma-retroviral and lentiviral (LV) delivery vectors to express a CD19-specific chimeric antigen receptor (CAR) for cancer treatment. This therapeutic approach has shown remarkable results against B-cell malignancies in pilot clinical trials. Consequently, there is a strong desire to make CAR T cell therapy scalable and globally available to patients. However, there are persistent concerns and limitations with the use of viral vectors for CAR T cell generation with regard to safety, cost and scale of vector production. In order to address these concerns, we aimed to improve non-viral gene transfer and genome editing tools as an effective, safe and broadly applicable alternative to viral delivery methods for T-cell engineering. In the first part of the study, we engineered CAR T cells through non-viral Sleeping Beauty (SB) transposition of CAR genes from minimalistic DNA vectors called minicircles rather than conventional SB plasmids. This novel approach dramatically increased stable gene transfer rate and cell viability and resulted in higher yield of CAR+ T cells without the need of long ex vivo expansion to generate therapeutic doses of CAR+ T cells. Importantly, CD19-CAR T cells modified by MC-based SB transposition were equally effective as LV transduced CD19-CAR T cells in vitro and in a murine xenograft model (NSG/Raji-ffLuc), where a single administration of CD8+ and CD4+ CAR T cells led to complete eradication of lymphoma and memory formation of CAR T cells after lymphoma clearance. To characterize the biosafety profile of the CAR T cell products, we did the most comprehensive genomic insertion site analysis performed so far in T cells modified with SB. The data showed a close-to-random integration profile of the SB transposon with a higher number of insertions in genomic safe harbors compared to LV integrants. We developed a droplet digital PCR assay that enables rapid determination of CAR copy numbers for clinical applications. In the second part of the study, we ablated expression of PD-1, a checkpoint and negative regulator of T cell function to improve the therapeutic index of CAR T cells. This was accomplished using non-viral CRISPR/Cas9 via pre-assemble Cas9 protein and in vitro-transcribed sgRNA (Cas9 RNP). Finally, we combined our developed Cas9 RNP tool with CAR transposition from MC vectors into a single-step protocol and successfully generated PD-1 knockout CAR+ T cells. Based on the promising results achieved from antibody-mediated PD-1 blockade in the treatment of hematological and solid tumors, we are confident that PD-1 knockout CAR T cells enhance the potency of CAR T cell therapies for treatment of cancers without the side effects of antibody-based therapies. In conclusion, we provide a novel platform for virus-free genetic engineering of CAR T cells that can be broadly applied in T-cell cancer therapy. The high level of gene transfer rate and efficient genome editing, superior safety profile as well as ease-of-handling and production of non-viral MC vectors and Cas9 RNP position our developed non-viral strategies to become preferred approaches in advanced cellular and gene-therapy.}, subject = {Krebs }, language = {en} } @phdthesis{Heisig2010, author = {Heisig, Martin}, title = {Development of novel Listeria monocytogenes strains as therapeutic agents for targeted tumor therapy}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-48628}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2010}, abstract = {Despite marked progress in development and improvement of cancer therapies the rate of cancer related death remained stable over the last years. Especially in treating metastases alternative approaches supporting current therapies are required. Bacterial and viral vectors have been advanced from crude tools into highly sophisticated therapeutic agents detecting and treating neoplastic leasions. They might be potent enough to fill in this therapeutic demand. In this thesis Listeria monocytogenes was investigated as carrier for targeted bacterial cancer therapy. One part of the study focussed on modification of a functional bacterial mRNA delivery system. Genomic integration of T7 RNA polymerase driving mRNA production allowed reduction to an one-plasmid-system and thereby partially relieved the growth retardation exerted by mRNA delivery. Importantly the integration allowed metabolic attenuation of the mRNA delivery mutant potentially enabling in vivo applications. Further expansion of the bacterial RNA delivery system for transfer of shRNAs was examined. Bacterial mutants producing high amounts of RNA containing shRNA sequences were constructed, however a functional proof of gene silencing on delivery in eukaryotic cell lines was not achieved. The second part of this thesis focussed on increasing tumor colonization by Listeria monocytogenes in vivo. Coating bacteria with antibodies against receptors overexpressed on distinct tumor cell lines enabled specific bacterial internalization into these cells in vitro. Optimization of the bacterial antibody coating process resulted in an up to 104-fold increase of intracellular bacteria. Combination of this antibody-mediated targeting with the delivery of prodrug-converting enzymes showed a cytotoxic effect in cell lines treated with the corresponding prodrug. Since incubation in murine serum completely abrogated antibodymediated bacterial internalization the antibodies were covalently linked to the bacteria for application in xenografted tumor mice. Bacteria coated and crosslinked in this manner showed enhanced tumor targeting in a murine tumor model demonstrating antibodymediated bacterial tumor targeting in vivo. Independent of antibody-mediated tumor targeting the intrinsic tumor colonization of different Listeria monocytogenes mutants was examined. Listeria monocytogenes \&\#916;aroA \&\#916;inlGHE colonized murine melanoma xenografts highly efficient, reaching up to 108 CFU per gram of tumor mass 7 days post infection. Taken together the presented data shows highly promising aspects for potential bacterial application in future tumor therapies. Combination of the delivery systems with antibodymediated- and intrinsic bacterial tumor targeting might open novel dimensions utilizing Listeria monocytogenes as therapeutic vector in targeted tumor therapy.}, subject = {Krebs }, language = {en} } @phdthesis{Junker2015, author = {Junker, Markus}, title = {Development and characterization of monoclonal antibodies to GDF-15 for potential use in cancer therapy}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-132424}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {Background GDF-15 is a divergent member of the TGF-superfamily, which was first described as macrophage inhibitory cytokine-1 (MIC-1), revealing an immune modulatory function. GDF-15 is a soluble protein which is, under physiological conditions, highly expressed in the placenta and found in elevated levels in blood sera of pregnant women. Apart from the placenta, GDF-15 is expressed in healthy tissue, albeit to a lower extent and overexpressed in many solid tumors. A variety of different functions are attributed to GDF-15 in healthy as well as diseased humans. On the one hand, GDF-15 is required for successful pregnancy and low GDF-15 serum levels during pregnancy correlate with fetal abortion. On the other hand, overexpression of GDF-15, which can be observed in several malignancies is correlated with a poor prognosis. Furthermore, tumor derived GDF-15 leads to cancer associated anorexia-cachexia syndrome in mice. The aim of my PhD thesis was to further investigate the role of GDF-15 as an immune modulatory factor in cancer, in particular, by inhibiting the target molecule in vitro and in vivo. Therefore, the main focus was placed on the generation and characterization of monoclonal GDF-15 specific blocking antibodies, which were tested in vitro and in vivo, which represents a substantial part of my work. Results Here, GDF-15 was shown to be highly expressed in human gynecological cancer and brain tumors. We could then demonstrate that GDF-15 modulates effector immune cells in vitro. GDF-15 mediated a slight downregulation of the activating NKG2D receptor on NK and CD8+ T cells, which is crucial for proper anti-tumoral immune responses. Furthermore, we could demonstrate that GDF-15 reduces the adhesion of CD4+ and CD8+ T cells on endothelial cells in vitro. A negatively affected trans-endothelial migration of leukocytes into inflamed tissue could explain the low T cell infiltration in GDF-15 expressing tumors, which were observed in vivo, where mice bearing (shRNA mediated) GDF-15 deficient glioma cells revealed enhanced immune cell infiltrates in the tumor microenvironment, compared with the GDF-15 expressing control group. Those animals further exhibited a decreased tumor growth and prolonged survival. GDF-15 is a soluble protein, secreted by more than 50 \% of solid tumors and associated with grade of malignancy. Therefore a neutralizing monoclonal antibody to GDF-15 was assumed to be an auspicious therapeutically anti-cancer tool. Such an antibody was thus generated in GDF-15 knock out mice against human GFD-15. Amongst many clones, the GDF-15 antibody clone B1-23 was found to be applicable in Western Blot as well as in ELISA techniques, detecting a three-dimensional epitope of the mature GDF-15 dimer with high affinity and specificity. To enable the humanization for a later administration in humans, the variable regions of antibody B1-23 were identified by a special PCR method using degenerate primers and cloned into a sequencing vector. The sequence obtained thereby enabled the generation of chimeric and humanized B1-23 variants. After further comprehensive characterization, the original mouse antibody B1-23 as well as the chimeric antibody (ChimB1-23) and the humanized B1-23 antibody (H1L5) were applied in a melanoma xenograft study in vivo. None of the antibodies could significantly inhibit tumor growth. .However of utmost importance, body weight loss mediated by tumor derived GDF-15 could be significantly prevented upon administration of all three GDF-15 specific antibodies, which confirmed the antagonizing functionality of the immunoglobulin. Conclusion GDF-15 is a promising cancer target, involved in tumor progression and cancer related cachexia. A monoclonal GDF-15 antibody was generated, which served on one hand as a tool for molecular biological applications (Western Blot, ELISA, etc.) and on the other hand was applied as an antagonizing antibody in vitro and in vivo. Even though tumor growth inhibition by GDF-15 depletion in T cell deficient athymic mice failed using B1-23, the same antibody and derivates thereof (chimeric and humanized) impressively prevented tumor associated cachexia in UACC-257 melanoma bearing nude mice. The missing anti-tumor effect in our own melanoma model in nude mice can only partially be explained by the missing secondary immunity, in particular cytotoxic T cells, in the athymic animals, since in a similar melanoma model, performed by an external company, a tumor reduction in immunocompromised animals was observed, when B1-23 was administered. These findings support the idea that T cells are substantial for an effective tumor immunity and are in line with the results of the syngeneic, T cell comprising, mouse glioma model, where silencing of tumor expressed GDF-15 led to an enhanced intratumoral T cell infiltration and a prolonged survival. Taken together our data allow for the conclusion that tumor associated cachexia can be combatted with the GDF-15 antibody B1-23. Further, B1-23 might elicit direct anti-tumor effects in immune competent models, which contain T cells, rather than in an athymic, T cell deficient nude mouse model.}, subject = {Growth-differentiation Factor 15}, language = {en} } @phdthesis{WeinstockgebPattschull2019, author = {Weinstock [geb. Pattschull], Grit}, title = {Crosstalk between the MMB complex and YAP in transcriptional regulation of cell cycle genes}, doi = {10.25972/OPUS-17086}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-170866}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2019}, abstract = {The Myb-MuvB (MMB) multiprotein complex is a master regulator of cell cycle-dependent gene expression. Target genes of MMB are expressed at elevated levels in several different cancer types and are included in the chromosomal instability (CIN) signature of lung, brain, and breast tumors. This doctoral thesis showed that the complete loss of the MMB core subunit LIN9 leads to strong proliferation defects and nuclear abnormalities in primary lung adenocarcinoma cells. Transcriptome profiling and genome-wide DNA-binding analyses of MMB in lung adenocarcinoma cells revealed that MMB drives the expression of genes linked to cell cycle progression, mitosis, and chromosome segregation by direct binding to promoters of these genes. Unexpectedly, a previously unknown overlap between MMB-dependent genes and several signatures of YAP-regulated genes was identified. YAP is a transcriptional co-activator acting downstream of the Hippo signaling pathway, which is deregulated in many tumor types. Here, MMB and YAP were found to physically interact and co-regulate a set of mitotic and cytokinetic target genes, which are important in cancer. Furthermore, the activation of mitotic genes and the induction of entry into mitosis by YAP were strongly dependent on MMB. By ChIP-seq and 4C-seq, the genome-wide binding of MMB upon YAP overexpression was analyzed and long-range chromatin interaction sites of selected MMB target gene promoters were identified. Strikingly, YAP strongly promoted chromatin-association of B-MYB through binding to distal enhancer elements that interact with MMB-regulated promoters through chromatin looping. Together, the findings of this thesis provide a so far unknown molecular mechanism by which YAP and MMB cooperate to regulate mitotic gene expression and suggest a link between two cancer-relevant signaling pathways.}, subject = {Krebs }, language = {en} } @phdthesis{Wolter2015, author = {Wolter, Patrick}, title = {Characterization of the mitotic localization and function of the novel DREAM target GAS2L3 and Mitotic kinesins are regulated by the DREAM complex, often up-regulated in cancer cells, and are potential targets for anti-cancer therapy}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-122531}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2015}, abstract = {The recently discovered human DREAM complex (for DP, RB-like, E2F and MuvB complex) is a chromatin-associated pocket protein complex involved in cell cycle- dependent gene expression. DREAM consists of five core subunits and forms a complex either with the pocket protein p130 and the transcription factor E2F4 to repress gene expression or with the transcription factors B-MYB and FOXM1 to promote gene expression. Gas2l3 was recently identified by our group as a novel DREAM target gene. Subsequent characterization in human cell lines revealed that GAS2L3 is a microtubule and F-actin cross-linking protein, expressed in G2/M, plays a role in cytokinesis, and is important for chromosomal stability. The aim of the first part of the study was to analyze how expression of GAS2L3 is regulated by DREAM and to provide a better understanding of the function of GAS2L3 in mitosis and cytokinesis. ChIP assays revealed that the repressive and the activating form of DREAM bind to the GAS2L3 promoter. RNA interference (RNAi) mediated GAS2L3 depletion demonstrated the requirement of GAS2L3 for proper cleavage furrow ingression in cytokinesis. Immunofluorescence-based localization studies showed a localization of GAS2L3 at the mitotic spindle in mitosis and at the midbody in cytokinesis. Additional experiments demonstrated that the GAS2L3 GAR domain, a putative microtubule- binding domain, is responsible for GAS2L3 localization to the constriction zones in cytokinesis suggesting a function for GAS2L3 in the abscission process. DREAM is known to promote G2/M gene expression. DREAM target genes include several mitotic kinesins and mitotic microtubule-associated proteins (mitotic MAPs). However, it is not clear to what extent DREAM regulates mitotic kinesins and MAPs, so far. Furthermore, a comprehensive study of mitotic kinesin expression in cancer cell lines is still missing. Therefore, the second major aim of the thesis was to characterize the regulation of mitotic kinesins and MAPs by DREAM, to investigate the expression of mitotic kinesins in cancer cell line panels and to evaluate them as possible anti-cancer targets. ChIP assays together with RNAi mediated DREAM subunit depletion experiments demonstrated that DREAM is a master regulator of mitotic kinesins. Furthermore, expression analyses in a panel of breast and lung cancer cell lines revealed that mitotic kinesins are up-regulated in the majority of cancer cell lines in contrast to non-transformed controls. Finally, an inducible lentiviral-based shRNA system was developed to effectively deplete mitotic kinesins. Depletion of selected mitotic kinesins resulted in cytokinesis failures and strong anti-proliferative effects in several human cancer cell lines. Thus, this system will provide a robust tool for future investigation of mitotic kinesin function in cancer cells.}, subject = {Zellzyklus}, language = {en} } @phdthesis{Polzien2011, author = {Polzien, Lisa}, title = {BAD Phosphorylation: A Novel Link between Apoptosis and Cancer}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-56919}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2011}, abstract = {BAD (Bcl-2 antagonist of cell death, Bcl-2 associated death promoter) is a pro-apoptotic member of the Bcl-2 protein family that is regulated by phosphorylation in response to survival factors. Although much attention has been devoted to the identification of phosphorylation sites in murine BAD (mBAD), little data are available with respect to phosphorylation of human BAD (hBAD) protein. In this work, we investigated the quantitative contribution of BAD targeting kinases in phosphorylating serines 75, 99 and 118 of hBAD (Chapter 3.1). Our results indicate that RAF kinases phosphorylate hBAD in vivo at these established serine residues. RAF-induced phosphorylation of hBAD was not prevented by MEK inhibitors but could be reduced to control levels by use of the RAF inhibitor Sorafenib (BAY 43-9006). Consistently, expression of active RAF suppressed apoptosis induced by hBAD and the inhibition of colony formation caused by hBAD could be prevented by RAF. In addition, using surface plasmon resonance technique we analyzed the direct consequences of hBAD phosphorylation by RAF with respect to complex formation of BAD with 14-3-3 proteins and Bcl-XL. Phosphorylation of hBAD by active RAF promotes 14-3-3 protein association, whereby the phosphoserine 99 represents the major binding site. Furthermore, we demonstrate in this work that hBAD forms channels in planar bilayer membranes in vitro. This pore-forming capacity is dependent on phosphorylation status and interaction with 14-3-3 proteins. Additionally, we show that hBAD pores possess a funnel-shaped geometry that can be entered by ions and non-charged molecules up to 200 Da (Chapter 3.2). Since both lipid binding domains of hBAD (LBD1 and LBD2) are located within the C-terminal region, we investigated this part of the protein with respect to its structural properties (Chapter 3.3). Our results demonstrate that the C-terminus of hBAD possesses an ordered β-sheet structure in aqueous solution that adopts helical disposition upon interaction with lipid membranes. Additionally, we show that the interaction of the C-terminal segment of hBAD with the BH3 domain results in the formation of permanently open pores, whereby the phosphorylation of serine 118 proved to be necessary for effective pore-formation. In contrast, phosphorylation of serine 99 in combination with 14-3-3 association suppresses formation of channels. These results indicate that the C-terminal part of hBAD controls hBAD function by structural transitions, lipid binding and phosphorylation. Using mass spectrometry we identified in this work, besides the established in vivo phosphorylation sites at serines 75, 99 and 118, several novel hBAD phosphorylation sites (serines 25, 32/34, 97, 124 and 134, Chapter 3.1). To further analyze the regulation of hBAD function, we investigated the role of these newly identified phosphorylation sites on BAD-mediated apoptosis. We found that in contrast to the N-terminal phosphorylation sites, the C-terminal serines 124 and 134 act in an anti-apoptotic manner (Chapter 3.4). Our results further indicate that RAF kinases and PAK1 effectively phosphorylate BAD at serine 134. Notably, in the presence of wild type hBAD, co-expression of survival kinases, such as RAF and PAK1, leads to a strongly increased proliferation, whereas substitution of serine 134 by alanine abolishes this process. Furthermore, we identified hBAD serine 134 to be strongly involved in survival signaling in B-RAF-V600E containing tumor cells and found phosphorylation of this residue to be crucial for efficient proliferation in these cells. Collectively, our findings provide new insights into the regulation of hBAD function by phosphorylation and its role in cancer signaling.}, subject = {Krebs }, language = {en} }