Unsere Geförderten

Im Folgenden möchten wir Ihnen unsere aktuell geförderten Ärzte/innen und Wissenschaftler/innen vorstellen und Ihnen einen kurzen Einblick in deren Fachgebiete und Projekte gewähren.

Wir möchten besonders junge Nachwuchskräfte (Doktoranden/innen der Medizin und Masterstudenten/innen) auf die Arbeitsgruppen und Projekte der Geförderten aufmerksam machen und freuen uns bei Interesse an einer Mitarbeit in einer der Gruppen über Ihre Kontaktaufnahme!

Bei Interesse für eine naturwissenschaftliche Doktorarbeit wenden Sie sich ebenfalls gerne an uns, wir stellen den Kontakt her und unterstützen Sie bei der eigenständigen Einwerbung einer Finanzierung (z.B. eines Doktorandenstipendiums).

Partnerlabore

  • DNA repair, hypoxia and genomic instability in metastatic prostate cancer (DRAGOON): Mechanistic insights and new treatment options

    Dissemination of tumour cells and metastatic spread as well as cancer resistance to standard treatment are still the common cause of treatment failure and eventually death in prostate cancer patients. A better understanding the biology of the above-mentioned features will indeed lead to more effective cancer therapy options in prostate cancer. To that end, molecular techniques mixed with scientific experience of the medical scientist Dr. Mansour complemented by clinical medical oncology expertise of Dr. Oing are gathered to promote the current bidirectional translational research project entitled DRAGOON. In DRAGOON, several high throughput techniques in various in vivo, in vitro and ex vivo models will be employed to address features of dissemination and metastasis as well as endocrine resistance in this context, by examining incurable prostate cancer at

    (i) an early (hormone-sensitive, node-positive) (subproject 1) and also (ii) a late (metastatic castration-resistant) (subproject 2) stage of metastatic disease as a special exemplar, taking into account the complexity of advanced prostate cancer. Both subprojects aim to identify distinct signatures within the genome, transcriptome, epigenome and/or kinome, which contribute to prostate cancer dissemination, metastasis and progression to castration resistant phenotype, which will assist in developing new biomarkers and novel, biomarker-driven, personalised therapeutic strategies (subproject 3).

    Contact and affiliation:

    PD Dr. rer. nat Wael Y. Mansour

    Laboratory for Radiobiology & Experimental Radiation Oncology
    w.mansour@uke.de
    +49 (0) 40 7410 56520
    Website: Regulation of DNA Double Strand Break Repair in Tumors

    Dr. med. Christoph Oing

    II. Medical Clinic and Polyclinic for Oncology and Hematology
    c.oing@uke.de
    +49 (0) 40 7410 55139

    NUAcT Fellow
    NU Translational & Clinical Research Institute
    CRUK NU Drug Discovery Unit
    Newcastle University

    Honorary Consultant Medical Oncologist Early Clinical Trials Unit
    Sir Bobby Robson Cancer Trials Research Centre
    Northern Centre for Cancer Care@Freeman Hospital
    Newcastle Hospitals NHS Foundation Trust

  • Dissecting the metastatic cascade to improve the clinical management of ovarian cancer

    Phenotypically and genetically heterogeneous tumors are more aggressive, have a higher relapse rate, and are more resilient to therapy. Metastatic outgrowth of disseminated tumor cells is thought to be fueled by clonal cooperation of tumor clones from early and late stages of cancer evolution. Circulating tumor cells (CTCs) can be detected in the peripheral blood and peritoneal liquid in most of the ovarian cancer patients in the different stages of the disease. Literature suggests that CTCs that found their way into the blood stream may in general not be able to extravasate and colonialize outside the peritoneal cavity, but may in fact return to the primary site to perform so called self‐seeding. The process of growing the tumor from the “outside in”maybe one of the mechanisms of maintaining tumor heterogeneity and has significant clinical implications. This study will elucidate the metastatic cascade of ovarian cancer by investigating the natural cancer evolution through the analysis of single tumor cells on genomic, transcriptomic, proteomic, and methylomic (multi‐omic) level. Dissecting the metastatic cascade in ovarian cancer and understanding the evolutionary pathways of how and when metastasis takes place will greatly help in the management of cancer patients.

    Contact and affiliation:

    PD Dr. med. Katharina Prieske

    Department of Gynecology
    k.prieske@uke.de
    +49 (0) 40 7410 54355

    Dysplasiezentrum am Krankenhaus Jerusalem
    Moorkamp 2-6
    20357 Hamburg
    040-44190-450
    Website

    PD Dr. Simon A. Joosse, PhD

    Department of Tumor Biology
    s.joosse@uke.de
    +49 (0) 40 741051970
    Website

  • The RESISTOME of metastatic medulloblastoma: Unravelling molecular mechanisms for personalized therapy

    Medulloblastoma (MB) is the most frequent high-grade (WHO IV) brain tumor of childhood and adolescence. Metastasis is one of the key clinical problems in MB. Multimodal therapy of MB consists of combined surgery, chemotherapy and radiotherapy. Radiothearpy probably is the most effective modality, but besides other effects, considerablely interferes with neurocognitive development especially in young children. Available treatment options may be associated with transient responses but often provide no cure. Response to therapy is affected by the intrinsic RESISTOME of tumor cells, which is related to molecular processes such as DNA repair, replication and stemness. The RESISTOME study will investigate the mechanisms underlying the response and resistance towards radiotherapy in MB. The study strives to introduce molecular targeting strategies to increase the radiosensitivity of primary MB and spinal metastases to enable personalized therapy and improve patient survival. In this bidirectional translational research project we will combine the expertise of the clinician scientist Dr. Martin Mynarek on pediatric oncology and clinical data analysis with the expertise of the medical scientist Dr. Nina Struve in experimental radiobiology and clinical radio-oncology to interpret data from in-vitro and ex-vivo models as well as patient data to address radio- and chemoresistance, molecular targeting, and response prediction in the context of MB.

    Contact and affiliation:

    Dr. rer. nat. Nina Struve

    Department of Radiobiology and Radiation Therapy/Reference Center for RT for MB & Pineoblastoma
    ni.struve@uke.de
    +49 (0)40 7410 59789/59413
    +49 (0)40 7410 59413

    Dr. med. Martin Mynarek

    Department of Pediatric Hematology & Oncology

    Children´s UKE
    m.mynarek@uke.de
    +49 (0) 40 7410 53394

Clinician Scientists

  • Histological, molecular and clinical characterization of MYB/MYBL1-altered gliomas

    Currently, only few data on gliomas with alterations of the MYB or MYBL1 genes are available. Thus, they are often misdiagnosed, the prognosis is unclear and targeted therapies do not exist. I will assemble a patient cohort of approximately 100 MYB/MYBL1-altered gliomas to define subgroups with cluster analyses of DNA methylation data. Next, I will analyse the histology and immunohistochemical profile of these gliomas to investigate whether the histology, and also MRI features, correlate with the molecular subgroups and differ from other glioma entities. These data will also give insight whether MYB/MYBL1-altered gliomas, especially those with a high proliferation, may show a wider dissemination and metastasize. RNA sequencing will identify gene fusions in these gliomas, reveal differential gene expression compared to other gliomas and unravel signalling pathways active in MYB/MYBL1-altered gliomas. I will then express the gene fusions detected in murine cell lines in vitro to study the individual effect on cell growth. All these data will lead to a better understanding of the tumor biology of MYB/MYBL1-altered gliomas, improve diagnostic pipelines while also integrating molecular data, and indicate therapeutic targets for clinical use. Finally, I will analyse clinical data of patients with MYB/MYBL1-altered gliomas to get insight into the prognosis.

    Contact and affiliation:

    Dr. med. Annika Wefers

    Institute of Neuropathology
    Phone: 0152-22827602
    Email: a.wefers@uke.de

  • Design and preclinical development of multi-specific nanobodies by targeting key players of thepurinergic pathway (CD39, CD73) in combination with TIGIT/PVR/PVRL2 axis for disseminatedhematological cancer (Acute Myeloid Leukemia (AML), Multiple Myeloma (MM))

    The aim of our project is to employ nanobody-based biologics to reactivate T cell immunity against disseminating hematological malignancies by targeting immunosuppressive checkpoint and adenosine pathways. Our expertises synergize ideally to achieve this goal: Stephan Menzel has developed inhibitory nanobodies (Nbs) for CD39 and CD73 and will perform Nb discovery against TIGIT, PVR and PVRL2. He will analyze Nb functions regarding enzymatic inhibition and Fc mediated effector functions. Franziska Brauneck will analyze the effects of blocking CD39 and CD73 alone or in combination with TIGIT-PVR blockade on T-cell activation and cancer cell killing in vitro and in mouse models. Nbs against ectonucleotidases and checkpoint inhibitors will serve as a basis for developing multi-specific inhibitors to modulate purinergic signaling and checkpoint inhibition as a novel anticancer strategy. Our in vitro results will guide us to analyze in vivo efficacy in established mouse models with the clear aim to be translated in a bench-to-bedside project into early clinical first-in-man application. We hope to benefit strongly from the excellent UCCH infrastructure, the MSNZ community and ongoing clinical work, expecting to contribute in-depth knowledge and new clinical tools to other projects in the field of dissemination and metastasis.

    Contact and affiliation:

    Dr. med. Franziska Brauneck

    Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology
    f.brauneck@uke.de
    +49 (0) 40 7410 50287


  • Identification of the soluble factor mediating CAF induced proliferation and everolimus resistance in PanNET – an experimental and translational study

    Most of patients with PanNET require systemic treatment during the course of disease. Up to 50% of patients present with metastatic disease at diagnosis and even after curative intended surgery for localized disease, 20-30% of patients experience a recurrence. Therapeutic options include the mTOR inhibitor everolimus. However, primary and secondary resistance upon everolimus treatment has not been well studied. Although CAF have been identified as relevant contributors to cancer progression and drug resistance in many tumors, no study has addressed whether CAF are involved in progression and therapeutic resistance in NET. In my doctoral thesis, I examined the in vitro interaction of CAF and PanNET(NT-3, BON cells) and could verify these results in human PanNET tissue. I demonstrated dedifferentiation, induction of proliferation and therapeutic resistance towards everolimus through STAT3-activation in PanNET cells upon stimulation with CAF. However, we have not yet identified the soluble factor(s) mediating these effects. The aim of this project is to identify the soluble factor mediating this stroma-tumor interaction and thereby define novel therapeutic targets to halt tumor growth and restore everolimus responsiveness in PanNET. During the funding period, experiments will be carried out using human PanNET cells, NT-3 and NT-18, and human CAF derived from liver metastases and primary tumors of PanNET. Furthermore, human tissue and serum samples from PanNET patients will be analyzed. Labspace and funding for consumables will be provided by the I. Department of Medicine lead by Prof. Lohse and Prof. Huber. We furthermore aim to submit a grant to the Deutsche Forschungsgemeinschaft (DFG) by end of the funding period.

    Affiliation and contact:

    Dr. med. Tania Amin

    Department of Gastroenterology
    t.amin@uke.de

  • A multimodal analysis of genomic patterns of relapse and dissemination in supratentorial ependymoma

    Ependymomas are malignant tumours of the central nervous system (CNS) which affect both children and adults and that can be stratified into at least ten molecular groups with heterogenous biological features and varying course of disease. To date, the mainstay of therapy for ependymoma remains maximal safe surgery and radiotherapy. Whereas some patients can be cured using this approach, high-risk subgroups relapse frequently, resulting in an unfavourable prognosis with significant morbidity and mortality. The knowledge about the genetic changes of ependymoma at relapse remains incomplete. In this project, we aim at systematically comparing pairs of primary and relapsed or disseminated supratentorial ependymoma (ST-EPN) to increase our understanding of the biological foundations of relapse and dissemination, and to define prognostic markers that may aid risk stratification in future trials. To this end, patient samples and clinical data will be systematically collected from the UKE biobank, through the HIT network and INFORM-study and subjected to a) a set of core methods to establish an integrated set of clinical and multi-omics data for each primary-relapse/dissemination pair, and b) advanced single nucleus RNA-sequencing (snRNA-seq.) and spatial transcriptomics methods. The external cooperation partner for this project will be the Hopp-Children’s Cancer Center (KiTZ) in Heidelberg.

    Affilation and contact:

    David Ghasemi

    Department of Pediatric Hematology and Oncology, UKE
    Research Institute Children's Cancer Center Hamburg
    d.noralighasemi@uke.de

  • Mechanisms and clinical implications of intratumoral heterogeneity in malignant peripheral nervesheath tumors

    Malignant peripheral nerve sheath tumors (MPNST) are highly aggressive tumors, characterized by early metastasis and a devastating prognosis. MPNST arise through malignant transformation of benign plexiform neurofibroma (PNF) via an intermediate state called atypical neurofibroma (ANF). So far, the exact mechanisms of malignant progression are not sufficiently understood. MPNST typically display a pronounced intratumoral heterogeneity, with areas of high malignancy and such corresponding to ANF or even PNF. In collaboration with the Neurofibromatosis Outpatient Clinic , will exploit this heterogeneity to reconstruct the exact mechanisms of malignant transformation from PNF to ANF and MPNST and to identify possible targeted therapies. Firstly, we will perform detailed morphological, epigenetic andctranscriptomic profiling of specific PNF and ANF as well as heterogeneous areas within MPNST. Second, we will investigate the corresponding tumor microenvironment (TME) by single-cell and spatially resolved transcriptome analysis to identify critical cell populations and their interaction with tumor cells during malignant progression. In parallel, we will determine optimal culture conditions for tumor slice cultures and 3D organoid models of primary MPNST in order to build an in-house platform for preclinical drug testing in a patient specific approach.

    Affiliation and contact:

    Dr. med. Catena Kresbach

    Institute of Neuropathology, UKE
    Department of Pediatric Hematology and Oncology, UKE
    Research Institute Children's Cancer Center Hamburg
    c.kresbach@uke.de

Medical Scientists - Kooperation

  • Evaluation of innovative antibody fragment based targeted constructs as first line or combination treatment option for medulloblastoma and rhabdomyosarcoma using an in vitro cell-based Blood-Brain-Barrier Technology Platform

    Partner: Dr. Ole Pless

    The paediatric solid tumours medulloblastoma (MB, brain tumour) and rhabdomyosarcoma (RMS, soft tissue tumour) show overexpression of the epidermal growth-factor receptor (EGFR) and mutations or abnormal signalling in the PI3-kinase-pathway, which can lead to disseminated tumour growth. Both cancers metastasize; in RMS brain metastasis can develop. Conventional radio-and chemotherapy often results in severe long-term side-effects in young patients, such as problems with growth, learning or brain development. Therefore, targeted treatment options are urgently needed. We previously developed targeted therapies using antibody fragments (scFvs), targeting the EGFR. EGFR-specific scFvs as part of immunoconjugates, namely bispecific T-cell engagers (BiTEs) and immunotoxins (ITs), kill selectively tumour cells without affecting healthy cells. Using them in MB or RMS patients with brain metastasis, they must cross the Blood-Brain-Barrier (BBB), a protective layer in the brain. To evaluate the diffusion kinetics of the components, we want to test them in an innovative in vitro-BBB-model obtained from the cooperation partner. EGFR overexpression and constitutive PI3-kinase signalling are connected with maintenance of cancer stem-cells. Consequently, both EGFR-immunoconjugates should be used in combination with a well-known PI3-kinase inhibitor with established BBB penetrance. Having functional scFv-immunotherapeutics available, alternative cancer stem-cell related target proteins can be tested by exchanging the scFvs.

    Contact and affiliation:

    Dr. rer. nat. Judith Niesen
    Research Institute Children’s Cancer Center Hamburg and Department of Pediatric Hematology and Oncology
    j.niesen@uke.de
    +49 (0) 40 426051214

    Dr. rer. nat. Ole Pless
    Institute for Molecular Biology and Applied Ecology at Fraunhofer IME Screening Port Hamburg
    ole.pless@ime.fraunhofer.de


  • Promotion of bone-seeking tumors through the mechanosensory network in bone

    Partner: Dr. Anton Davydok, Helmholtz-Zentrum Geesthacht Zentrum für Material- und Küstenforschung/DESY

    Bone is a highly dynamic tissue undergoing a constant matrix remodeling activity regulated by mechanical stimuli. When breast and prostate cancers metastasize to bone, a favored site, the fine-tuned bone remodeling can hijacked while fragility fractures increase. Yet, the crucial role of the mechano-regulated bone turnover is still to be determined in the context of bone metastases. In this project we aim to assess the role of the mechanosensory lacuno-canalicular network of osteocytes on tumor progression with bone metastasis. We have designed an interdisciplinary project utilizing high-resolution imaging techniques, including synchrotron radiation-based small- and wide- angle X-ray scattering (SAXS/WAXS), to analyze the local processes whereby impaired local bone quality contributes to tumor progression.

    Contact and affiliation:

    Dr. rer. nat. Katharina Jähn-Rickert
    Heisenberg Research Group Department of Osteology and Biomechanics
    University Medical Center Hamburg-Eppendorf
    Lottestr. 55a 22529
    Hamburg Germany
    Mail: k.jaehn@uke.de
    Phone: 0049 (0) 40 7410 26301

    Dr Anton Davydok,
    Institut für Werkstoffphysik, Helmholtz-Zentrum Hereon
    Out-staion DESY Notkestraße 85,
    22607 Hamburg
    www.gems.hereon.de

  • Multiplexed Detection of Circulating Tumor Cells in Prostate Cancer Using Nanotechnology Platforms

    Partner: Dr. Neus Feliu, Fraunhofer Institut for Applied Polymer Research (IAP), Center for Applied Nanotechnology (CAN)

    Dissemination of prostate cancer (PCa) cells to secondary sites has a significant impact on the formation of tumor metastasis and the fate of a patients’ course of the disease. It has been shown that the number and phenotypic heterogeneity of circulating tumor cells (CTCs) correlate with malignancy and prognosis. Liquid biopsies (LB) can non-invasively provide a real-time picture of a patient’s cancer by detecting CTCs. We propose a nanoparticle-based LB assay that combines the enumeration of CTCs and their characterization. After enumeration, the heterogeneous CTCs are analyzed with a library of fluorescent nanoparticles (fNPs). Each kind fNPs (with distinct emission wavelength) is biofunctionalized with PCa-specific biomarkers allowing the phenotyping of CTCs by the fluorescence signal. Enumeration and characterization occur in a single step with fluorescence analysis with a flow cytometer resulting in a cost-effective, time-efficient (<2 hours) LB assay. The developed multiplexed sensing platform will allow for the diagnosis and prognosis of PCa, providing also predictive information, treatment monitoring, and possibly early diagnosis of metastasis and recurrences. Furthermore, the LB assay is transferable to other tumor types by functionalizing the fNPs with the respective tumor-specific biomarkers.

    Contact and affiliation:

    Dr. Marina Mutas
    Department of Tumor Biology, UKE
    and
    Fraunhofer IAP CAN
    marina.mutas@uke.de

    Dr. Neus Feliu Torres
    Fachbereich Physik der Universität Hamburg
    Fraunhofer Institute for Applied Polymer Research
    Center for Applied Nanotechnology
    nfeliu@physnet.uni-hamburg.de

  • Investigating the impact of tandem motifs-induced CtBP polymerization using pre-clinical and clinical samples

    Gene transcription is strictly controlled by assemblies of transcription factors, coregulators and chromatin modifiers. CtBPs are a group of corepressors whose overexpression in tumors is associated with cell proliferation, migration and increase in EMT. In a joint collaboration effort between UKE/EMBL, we show that CtBP polymerization is induced by RAI2, an intrinsically disordered tumor suppressor protein through two identical tandem motifs leading to the relief of the CtBP corepressor activity. This mode of CtBP inactivation using tandem motifs has implications in cancer therapy and preventing disease progression. Initially, the overall structure and composition of the CtBP corepressor complex will be understood followed by investigating the molecular basis of toppling of corepressor complex using tandem motifs-containing protein/peptides using cell biology and structural biology techniques. The feasibility of the tandem PxDLS motif peptide for CtBP inactivation will be tested using established tumor biology techniques in prostate cancer cell lines. Finally, the CtBP polymerization will be investigated in a clinical setup using patient-derived tumor organoids and circulating tumor cells from advanced prostate cancer patients. The major aim here is to establish and validate tandem-motif induced CtBP polymerization as a therapeutic concept for a peptide-based inhibitor in cancer treatment.

    Affiliation and contact:

    Dr. rer. nat. Nishit Goradia

    Institute for Tumor Biology
    n.goradia@uke.de

  • High Precision Multiomics for Translational Cancer Research: µOmix4CaRe Micro-omics for cancer research

    In this project, we want to adapt an early stage prototype of a nanosecond infrared laser (NILR) tissue sampling device (3D MiTi LAb) to translational cancer research in collaboration with the UCCH. For this purpose, we need to develop a technical and methodical interface to integrate our system into the typical tissue and cell biology workflow. To achieve this aim, methods and tools development are required to register offline imaging data (e.g. microscopy data of stained tissue slices histologies) or lipid maps from MALDI imaging) to the 3D MiTi LAb coordinate system and use it as guidance for laser sampling. Furthermore, sample preparation of NIRL obtained tissue samples will be optimized with respect to the requirements of the specific omics technique. To adapt this technique to cancer research,we will build on our experience through preliminary studies with several collaborators and their cancer topics. Starting with prostate cancer as a model entity, we will evaluate our proposed approach workflow with histological and functional assays. To achieve this, a methodical biomedicine interface will fill the need to connect biomedical aspects with advanced analytical technologies, represented by a technical omics interface. We believe that gathering the research expertise of the partner laboratories (AG Schlüter and AG Mansour) will foster our seek for unfolding mechanisms associated with tumor initiation, heterogeneity, tumor microenvironment and metastatic development.

    Affiliation and contact:

    Dr.-Ing. Jan Hahn

    Institute for Clinical Chemistry and Laboratory Medicine
    ja.hahn@uke.de

  • “EUROPa” Exploring & Understanding Radiosensitivity of Pancreatic Cancer

    Pancreatic cancer (PDAC) is one of the most lethal cancers worldwide with early metastasis and chemoresistance. The stroma-enriched, immunosuppressive tumour microenvironment plays a keyrole in PDAC development, aggressiveness and therapy resistance. Due to these complex conditions, PDAC therapy has largely remained static over the last decades, demanding new therapy approaches. According to its local as well as systemic effects, radiotherapy can be an – so far – unexploited but promising treatment option for advanced and also metastatic PDAC. By combining innovative, translational experimental systems (e.g. ex vivo tissue slice cultures, advanced in vitro co-cultures) with the joint expertise of the cooperation partners in the fields of radiobiology, experimental oncology, cancer immunology and tumour biology this project aims to deliver the biological basis for the stratification of PDAC patients who will possible benefit from radiotherapy.

    Affiliation and contact:

    Dr. rer. nat. Sabrina Köcher

    Laboratory for Radiobiology and Experimental Radiation Oncology
    s.koecher@uke.de

  • Targeted Nanoparticle-Based Isolation of Circulating Tumor Cells for Enhanced Metastatic Potential Assessment

    Metastasis is a major challenge in cancer therapy, emphasizing the need for improved methods to assess the metastatic potential of circulating tumor cells (CTCs). In this study, we propose a novel approach utilizing targeted nanoparticles to isolate CTCs from blood samples, enabling a comprehensive evaluation of their metastatic potential. Key aspects are the specific targeting of the interaction between tumor cells and von Willebrand factor (vWF) as well as their adhesion ability to endothelial cells, to allow the isolation of CTCs with distinct properties. This research project encompasses nanoparticle design, synthesis, and characterization, followed by validation through mouse model studies and analysis of patient samples. The outcomes of this study have the potential to improve CTC detection and provide valuable insights for improved cancer diagnosis, prognosis, and personalized treatment strategies, ultimately enhancing patient outcomes in the field of oncology.

    Affiliation and contact:

    Dr. rer. nat. Yuanyuan Wang

    Department of Dermatology and Venereology
    yu.wang@uke.de

Short Term Clinician Scientists (Kurzzeit-/Anschubförderung)

  • Immune determinants of melanoma metastasis to the brain

    Despite recent advances in the treatment of advanced melanoma, MBMs remain a therapeutic challenge. Tothis day, the mechanisms of much worse prognosis for patients with MBMS than for patients with non-cranialmetastases remain unclear. We hypothesize that a less efficient anti-tumor immune response to MBMsaccounts for these different outcomes and propose to examine the systemic immunophenotype and localimmune reactions. Using existing blood samples from established repositories at the UKE, we plan on usingflow cytometry to describe the peripheral immune phenotypes of patients with MBM and compare these topatients with other distant melanoma metastases excluding the brain. Using existing tumor samples, we planon using regular immunohistochemistry, multiplex immunofluorescence and T-cell receptor sequencing toexamine the local anti-tumor immune response. This project proposal is a continuation of my previous researchand thesis work on primary brain tumors in the Department of Neurosurgery and establishes a newcollaboration between the Departments of Dermatology, Neurosurgery and Pathology.

    Contact and affiliation:

    Dr. med. Alessandra Rünger
    Department of Dermatology and Venerology
    In collaboration with “Labor für Hirntumorbiologie” – Department of Neurosurgery
    a.ruenger@uke.de

  • Immune Profiling for Individualized Treatment Decision in Diffuse Large B-Cell Lymphoma

    Patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) benefit from the introduction of immunotherapies. Simultaneously, conventional chemotherapy remains a crucial treatment component. The complex interplay and the possible implications for treatment decisions is poorly understood. We plan to conduct repeated blood sampling at important clinical hallmarks from 25 DLBCL patients during second-line treatment.In collaboration with Professor Eva Tolosa from the Department of Immunology we will design a comprehensive flow cytometry panel and functional tests to provide a longitudinal immune map of this cohort. This analysis will help us implement immunotherapies at the ideal time and avoid treatment failure. The long-term goal of this project is the development of a comprehensive immune analysis platform to predict treatment outcome of immunotherapies in lymphoma patients. The immunological focus of the University Medical Center Hamburg-Eppendorf provides the optimal environment to identify additional partners with specific knowledge or methods for this purpose. Simultaneously, an application for initial funding covering material resources is submitted to the “Hamburger Krebsgesellschaft”. The targeted start date is August 1st, 2024, with a continuous allocation of50% for clinical time and 50% for research time over a period of 12 months.

    Affilation and contact:

    Dr. med. Paul Kachel

    Department of Oncology and Hematology
    p.kachel@uke.de

  • Machine Learning Methods for the Integrated Molecular Classification of Ependymomas Using Histological Images, Clinical Characteristics and OMICS Data

    Ependymomas exhibit unique diagnostic challenges, e.g., data paucity and partially unclear relationship between molecular (DNA methylation) profiles and histological characteristics. Recently, neural networks were demonstrated to classify ependymoma types based on histology images, identifying morphological patterns associated with each tumor type (1). Typically, such networks are trained using weakly-supervised classification algorithms that process histological whole-slide images as smaller sub-images. This approach can be combined with clinicopathological and OMICS data, such as DNA methylation or proteomic profiling, for a more specific diagnosis. The aim of this project is to improve the generalizability and accuracy rate of a histology-based ependymoma classifier by optimization of input features and data augmentation. Diagnostic quality is further enhanced by combination with other data modalities, thus facilitating improved robustness and consistency. The integrated data is leveraged to analyze key regulators and their signaling pathways to prospectively allowing for targeted therapies of ependymoma subtypes. Overall, the objective of this project is to enhance the quality and consistency of ependymoma diagnoses with a machine-learning based approach that can be prospectively extended towards other brain tumor entities.

    Affiliation and contact:

    Dr. med. Maximilian Middelkamp

    Department of Neurosurgery
    m.middelkamp@uke.de

  • PD-L1 positive extracellular vesicles derived from exhaled breath condensate as a biomarker for lung cancer

    The high frequency and lethality of lung cancer create an urgent medical need for biomarkers to detect and monitor its progression. PD-L1+ EVs in the blood of lung cancer patients have been identified as cancer-derived particles. These vesicles have been found to directly correlate with tumor burden and may have the potential to differentiate between cancer patients and healthy individuals. Exhaled breath condensate (EBC) is a non-invasive fluid that can be easily obtained and may have higher diagnostic accuracy than blood due to its lower complexity. The aim of this study is to analyze the presence of PDL1+EVs in the EBC of patients with lung cancer and healthy controls using a super-resolution microscope (ONI Nanoimager). The primary endpoint of the analysis will be the presence and quantity of PD-L1+EVs in the EBC of lung cancer patients compared to healthy individuals. In addition, we plan to characterise other possible EV subtypes secreted from lung cancer cells. This initial exploratory study will form the basis for further studies. Subsequently, we aim to investigate whether the determination of PD-L1+ EVs from EBC is suitable for the early detection of lung cancer in high-risk populations. In addition, we aim to evaluate the method's applicability as a biomarker for follow-up during or after therapy.

    Affiliation and contact:

    Dr. med. Benjamin Schmidt

    Department of Oncology and Hematology
    ben.schmidt@uke.de

  • AURKA and ATR inhibition a novel targeted therapies in ARID1A mutated neuroendocrine neoplasms

    GEP-NEN are a rare and very heterogenous group of tumors including well differentiated neuroendocrine tumors (GEP-NET) and poorly differentiated neuroendocrine carcinomas (GEP-NEC). The latter respresents an extremely aggressive tumor diseases with a 5-year survival rate of approximately 25 %. Unfortunately, systemic therapy options for high grade GEP-NET and GEP-NEC are limited. So far, systemic chemotherapy with temozolomide/capecitabine or platin/etoposide is the only established therapeutic option. However, overall response rates are disappointing, and the majority of responsive patients will develop therapy resistance and disease progression. Therefore, new therapeutic strategies are urgently needed. The inactivating ARID1A mutation can be detected in approximately 40 % of cases of high grade GEP-NEN. Previous studies have shown that AURKA inhibition leads to selective synthetic lethality of ARID1A-deficient colorectal tumor cells. In our recent study, we were able to establish five novel GEP-NEN cell lines. Panel sequencing detected ARID1A mutation in GEP-NEC cell line NT-38 rendering them vulnerable to AURKA and ATR inhibition. Therefore, the aim of the present study is to further characterize AURKA and ATR inhibition in ARID1A mutated GEP-NEN and evaluate a liquid biopsy approach for the detection of ARID1A mutation in ctDNA. The obtained results could provide the basis for the first mutation based targeted therapy in high grade GEP-NEN.

    Affiliation and contact:

    Fabrice Viol M.Sc.

    Department of Gastroenterology
    f.viol@uke.de

Alumni

  • FrailPANC – Neoadjuvant treatment in frail patients with pancreatic adenocarcinoma – A prospective, randomized multi-centre Phase II AIO/ CHIR-Net Trial

    The majority of patients with pancreatic adenocarcinoma (PDAC) presents itself around 70 years, often in a frail physical condition (ECOG ≥2). Still, most clinical studies neglect this important patient subgroup. The aim of the FrailPANC trial is to evaluate the role of neoadjuvant chemotherapy in frail patients with resectable or borderline resectable PDAC. The primary endpoint is the overall survival, followed by the overall treatment utility (OTU) as a co-primary endpoint. In order to enable the trial a short project proposal will be submitted to the Deutsche Forschungsgemeinschaft (DFG) by February 2020. If the proposal is positively reviewed, the full proposal will be prepared within the requested funding period of the MSNZ (estimated start 06/2020). During the funding period the recruitment of participating sites and the integration of translational projects will be addressed. One of the translational projects will be in cooperation with the Department of Tumour Biology at the University Medical Center Hamburg-Eppendorf (UKE) with regard to their expertise concerning circulating tumour cells, single cell analytics and establishment of tumour organoids in cooperation with the Dutch Pancreatic Cancer Study Group. A second translational project will be established with the Department of Anaesthesiology regarding their preliminary work within the perioperative care of the elderly patient. In case of rejection of the proposal by the DFG, we will submit a grand proposal to the Deutsche Krebshilfe (DKH) during the MSNZ funding period.

    Contact and affiliation:

    Dr. med. Mara Goetz
    Department of General, Visceral and Thoracic Surgery
    m.goetz@uke.de
    +49 (0) 40 7410 54403

  • Studying the impact of tumor microenvironment and intestinal microbiota on CD4+ IL17+ heterogeneity in order to increase immunotherapy efficacy in colorectal cancer

    Recently, immunotherapies targeting co-inhibitory receptors (CIRs) have revolutionized the treatment of many types of cancer. However, in colorectal cancer (CRC), the third most common cancer, immunotherapies did not meet the expectations. We hypothesize that two key factors contribute to the poor efficacy of the immunotherapy in CRC: (I) the functional heterogeneity of the targeted cells, (II) the local microenvironment. Our unpublished data show that non-conventional IL17A+ Foxp3+ CD4+ T cells promote tumor growth and express several CIRs. This has to be taken into consideration since this population of protumorigenic cells can be unleashed by immunotherapies obtaining the opposite effects: growth of tumors. By characterizing this pro-tumorigenic cell population, we seek to determine the best immunotherapy regimen which is able to selectively impair the activity of these cells in CRC patients. Furthermore, we seek to understand in what manner the composition of tumor invading intestinal microbiota shapes the expression of CIRs on these cells and thereby, directing the efficacy of immunotherapy.

    Contact and affiliation:

    Dr. med. Leonie Konczalla
    Department of General, Visceral and Thoracic Surgery
    l.konczalla@uke.de
    +49 (0) 152 228 27 721

  • MATEO: Maintenance therapy vs. observation in FOLFIRINOX treated metastatic pancreatic ductal adenocarcinoma (mPDAC) patients - A prospective, randomized multi-center phase IIl AIO trial

    For initiation of the project, a study synopsis will be finished with a description of objectives and endpoints, study design, methodology and statistical considerations. This synopsis will serve as a first proposal submission at DFG. Further preparations for the final grant applications include preparation of relevant study documents (protocol, patient information material, ethics approval), recruitment of study centers within the AIO (Arbeitsgemeinschaft Internistische Onkologie) and initiation of the organization and management of the trial. Additionally, the preparation of a trials in progress manuscript for a medical journal is planned. Complementing, the development of a concomitant translational project is planned with utilizing the ideal starting point of this study for translational research with a homogenous study cohort, prospective collected data and design with a treatment and non-treatment arm. We plan to monitor therapy responses with assessment of circulating tumor markers (CA 19-9) and circulating tumor DNA (ctDNA) as possible therapy predictors along with the routine clinical and radiological assessments to establish subgroups for future therapy choices. I personally will conduct the described preparation of the grant application and organization of the trial with its accompanying translational program with PD Dr. Marianne Sinn as a supervisor and leading principal investigator of the study.

    Contact and affiliation:

    Dr. med. Martin Schönlein
    II. Medical Clinic and Polyclinic for Oncology and Hematology
    m.schoenlein@uke.de
    +49 (0) 152 228 72 093

  • Kinase activity profiles as prognostic marker for immunotherapy in head and neck squamous cell carcinoma

    Immune checkpoint inhibitors (ICI) have already been approved as 1st and 2nd line therapy of relapsed or distant metastatic head and neck squamous cell carcinoma (HNSCC). However, only 13-18% respond to therapy with no definitive predictive markers available resulting in a great need to establish predictive tests identifying responding and non-responding patients. For ICI therapy of advanced melanoma it has been demonstrated, that specific kinase activity clusters of peripheral blood mononuclear cells (PBMC) can be predictive. These clusters result from functional kinome profiling and might be promising in HNSCC. To test this, liquid biopsies will be collected from HNSCC patients before and under ICI treatment. PBMC will be isolated and functional kinome profiling will be performed. After cluster analysis the kinase activity clusters of the PBMC will be compared with the survival data of the patients. This method may also allow to monitor the patients` individual response during ICI treatment enabling the early detection of a progresses. Such a predictive test would support to identify responding versus non-responding patients, guide non-responders to a more appropriate therapy. This enables a more efficient economical use of the expensive therapeutics. In the future, a predictive test may also be transferred to additional entities.

    Contact and affiliation:

    Dr. med. Lara Bußmann
    Department of Otorhinolaryngology
    l.bussmann@uke.de

  • Cholangiocarcinoma complicating PSC – which T cells are promoting cancer dissemination?

    Cholangiocarcinoma (CCA) is a dreadful disease with a poor prognosis due to lacking therapeutic options, leading to death in 80 – 100 %. In Primary Sclerosing Cholangitis (PSC), a chronic and progressive inflammatory biliary disease, up to 40% of the patients develop CCA. The pathogenesis is poorly understood, but there is evidence that the immune system contributes towards promoting CCA. Histological studies have shown biliary intraepithelial metaplasia and dysplasia in PSC livers, suggesting an inflammation-driven metaplasia-dysplasia sequence CCA development Additionally, altered composition of the biliary microbiota from patients with PSC has been shown. The impact of the microbiota on T cell function - determined by the secretion of cytokines and expression of coinhibitory receptors - has been implicated in carcinogenesis. Thus, we hypothesize that protumorigenic T cells promote CCA development and dissemination. In order to define the functional phenotype, the co-inhibitory receptor profile, the origin of tumor infiltrating T cells and their interaction with tumor and biliary epithelial cells, we will perform single cell sequencing from blood, liver and tumor infiltrating T cells in combination with interactome analysis. Results will contribute to the understanding of the pathogenesis and reveal new immunotherapeutic options to directly serve patients suffering from this dreadful disease.

    Contact and affiliation:

    Dr. med. Jenny Krause
    I. Medical Clinic and Polyclinic
    je.krause@uke.de


  • Intratumoral heterogeneity of cancer stem cell features in hepatocellular carcinoma are decisive for indication of adjuvant treatment

    The genomic and histopathological intratumoral heterogeneity of HCC (hepatocellular carcinoma) is of increasing translational interest. Dismal prognosis is frequently linked to high invasiveness and metastatic potential of HCC harboring CSC-features, indicated by aberrant EpCAM-expression. However, to what extent intratumoral distribution of CSC-features impacts on recurrence after curative treatment remains unknown. We hypothesize that only homogeneous positivity will lead to higher invasiveness, early dissemination and dismal outcome, identifying patients in urgent need for adjuvant treatment. Analog to colorectal cancer, high or low risk features for recurrence could be decisive for adjuvant treatment. This proposal aims to investigate the spatial heterogeneity of EpCAM-expression within HCC-nodules and its impact on a) local invasiveness, b) early dissemination, and c) clinical outcome measured by time-to-recurrence (TTR) and recurrence-free-survival (RFS). Complementary, we aim to decipher the mutational background of CSC-features, addressing the mutational profile of the most frequent oncogenic drivers1,2 and focusing on the non-canonical role of telomerase on Wnt/β-catenin signaling through activating TERT-promoter mutations. For our preliminary experiments we already designed a heterogeneity-TMA of 82 patients (341 spots). EpCAM-staining, data on CTC-detection and serum levels of AFP, AFP-L3, DCP as well as marker for Wnt/β-catenin activation have been completed. The mutational profile is expected for 01/2020.

    Contact and affiliation:

    Dr. med. Kornelius Schulze
    I. Medical Clinic and Polyclinic
    k.schulze@uke.de


  • Unraveling resistance mechanisms to immune-checkpoint inhibitors via molecular characterization of CTC in metastatic melanoma patients

    Immune-checkpoint blockade therapy has revolutionized the treatment of patient with unresectable malignant melanoma. However, clinical outcome of the patients is obstructed by limited response rates and immune-related severe side effects. Therefore, there is an urgent but still unmet need for the identification of predictive biomarkers of clinical response. The aim of this project is to isolate circulating tumor cells from peripheral blood of metastatic melanoma patients treated by immune checkpoint inhibitors via a new enrichment method and to characterize them at the DNA and RNA level. Results from enumeration and molecular characterization will be correlated to clinical outcome and to other biological parameters in order to unravel diverse resistance mechanisms to improve the therapy regime. As melanoma is used as the prime model cancer entity for immunotherapy, this project will open new avenues for minimal invasive, blood-based, “liquid biopsy” biomarkers in other tumor entities (e.g., lung cancer) for which immunotherapy has also become a therapeutic option. This project is part of the LiquiMel Biobank which is organized by the applicant Julia Stadler and is a continuum of the UCCH fellowship of 2019. A close collaboration between the department of Dermatology and Tumorbiology supports the project.

    Contact and affiliation:

    Dr. med. Julia-Christina Stadler
    aktuell: Klinik für Dermatologie
    Medizinische Hochschule Hannover

    stadler-j@outlook.de


  • Unraveling tumor dormancy in humans by autopsies and post mortem biopsies

    In the cooperative research project between the Institute of Tumor Biology (ITB) and the Institute for Legal Medicine of the University Medical-Center Hamburg-Eppendorf (UKE), under the direction of Prof. Klaus Pantel and Prof. Klaus Püschel, I want to examine two scientific questions:

    1. What is the percentage of the population carrying dormant tumor cells at the time of death without being diagnosed with cancer ? This would provide an estimate about the rate of the occult early dissemination of tumor cells before cancer diagnosis. Routinely obtained (anonymized) bone marrow as well as peripheral blood samples collected from deceased persons at the risk age of developing cancer (> 45years) will be analyzed for the presence of disseminated tumor cells (DTCs) and circulating tumor cells (CTCs).

    2. Why are distant metastases in the skeletal muscles so rare even in patients with advanced cancer? Are CTCs not able to extravasate into muscle tissue or do DTCs exist in muscle tissue but remain in a dormant stage unable to grow out ? To investigate homing of CTCs to skeletal muscle (e.g. M. iliopsoas) originating from carcinoma patients, muscle samples of metastatic cancer patients (incl. solid tumors: gastro-intestinal and urogenital cancers, NSCLC and breast cancer, etc.) will be obtained during autopsies.

    Contact and affiliation:

    Dr. med. Antonia Fitzek
    Institute for Legal Medicine
    a.fitzek@uke.de
    +49 (0) 40 7410 - 57840

  • Generation of an anti α and β chain chimeric antigen receptor (αβ CAR) to develop CAR-T cells and CAR-NK cells for therapy of T cell leukemia

    Acute lymphoblastic leukemia (ALL) is the most common cancer in children. Even though ALL has a good overall prognosis, the more aggressive subgroup of relapsed T cell leukemia displays a poor outcome. Despite improvements in intensive chemotherapy and allogeneic hematopoietic stem cell transplantation (allo-HSCT), up to one third of pediatric T-ALL patients die of relapse or treatment failure. Therefore, new therapy options are urgently needed. In the last decades, immunotherapy became a turning point in cancer therapy. Antibodies targeting specific tumor antigens allow a highly specific and less toxic therapy. Moreover, cytotoxic immune cells modified with chimeric antigen receptors are able to kill tumor cells efficiently. Therefore, we aim to develop a chimeric antigen receptor targeting the alpha and beta chain of T cell receptors (αβTCR-CAR) to specifically destroy malignant T cells. Fratricide of CAR-T cells will be prevented by targeted disruption of the TCRβ locus.

    Contact and affiliation:

    Dr. med. Kerstin Schütze
    Department of Pediatric Hematology and Oncology

  • The impact of hypo-fractionated locally ablative radiotherapy, surgery, chemotherapy, and radio-chemotherapy on the development of metastases in two prostate cancer xenograft tumor models

    It is well known that radiotherapy can locally eliminate cancer cells. In addition to the local effects, radiotherapy also eliminates distant cancer cells, the so-called abscopal effect. However, its pathophysiological basis is not well understood and there is no information available on how to induce abscopal effects of radiotherapy that also influence the numbers of circulating tumor cells (CTCs) and disseminated tumor cells (DTCs). In recent experiments with two small cell lung cancer xenograft tumor models we could demonstrate that hypo-fractionated high dose radiotherapy can induce such abscopal effects lowering the numbers of CTCs and DTCs. But the mechanism underlying these observations are very poorly understood. Hence, we want to expand our experimental database with different prostate cancer xenograft models to investigate whether abscopal effects can also be seen in this additional clinically important tumor entity and how cells of the innate immune system are influenced by radiation.

    Contact and affiliation:

    PD Dr. med. rer. nat. Thorsten Frenzel
    Outpatient Center and Department for Radiotherapy and Radiation Oncology
    +49 (0) 40 7410 54031
    frenzel@uke.de

  • Relevance of SWI/SNF chromatin remodeling complex for gene expression and for regulation of transcription factors in human cancer - its role in the pathogenesis of acute myeloid leukemia as a model of malignant transformation in general -

    The aim of this project is to characterize the relevance of the so called SWI/SNF complex for gene expression and for regulation of transcription factors in human cancer. This complex is a chromatin remodeler, consisting of about 20 different proteins, acting as a destabilizer of histone-DNA interactions and thereby changing transcription activity of several genes. SMARCA4 (SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4, synonym: BRG1) is the central ATPase containing enzyme and of upmost interest for this project because it is well known to mediate tumor suppressive effects in solid tumors. However, in the setting of acute myeloid leukemia (AML), the basic field of research of this project, SMARCA4 seems to play a different role, acting as a proliferation stimulus for myeloid blasts making it an interesting target for new therapeutic approaches. The examination of its function via in vitro and in vivo analysis in combination with clinical data assessment in hematopoiesis and pathogenesis of AML could give deeper insights into involved pathways and SWI/SNF mediated function in tumorigenesis, proliferation and dissemination. AML could serve as a model for malignant transformation in general, extending the gained data to several other tumor entities.

    Contact and affiliation:

    Dr. med. Franziska Modemann

    II. Medical Clinic and Polyclinic for Oncology and Hematology
    f.modemann@uke.de


  • Characterization of the liquorigenic seeding and identification of metastatic risk factors in medulloblastoma with morpho-molecular profiling and machine learning

    Partner: Prof. Dr.- Ing. Andreas Schlaefer, TU Hamburg

    Medulloblastoma is the most frequent malignant pediatric brain tumor. Metastatic dissemination occurs primarily via the cerebrospinal fluid (CSF). Accurate detection of metastases including microscopic disease in the CSF is key for an adequate therapy stratification. However, this is a difficult and time-consuming task and cytological examination is frequently unreliable. Here, we propose to establish a machine learning pipeline to automatize and improve diagnostic evaluation of CSF samples. Further, we will perform an integrated morpho-molecular analysis to study the relation between morphological features of tumor cells in the CSF and molecular as well as clinical properties of the primary tumor.

    Contact and affiliation:

    Dr. med. Michael Bockmayr,
    Research Institute Children’s Cancer Center Hamburg and Department of Pediatric Hematology and Oncology
    +49 (0) 40 7410 52725
    m.bockmayr@uke.de

    Prof. Dr.-Ing. Andreas Schlaefer
    TU Hamburg, Medical Technology Science Systems

    schlaefer@tuhh.de

  • Antiresorptive Drug-Related Osteonecrosis of the Jaw following systemic treatment for bone metastases – characterizationby Raman spectroscopy, histomorphometry and quantitative backscattered electron imaging

    Antiresorptive agents such as bisphosphonates or the monoclonal RANKL-antibody denosumab are commonly administered in the treatment of bone metastases of solid tumors or multiple myeloma. They halt osteolysis by inhibiting osteoclast activity. A severe adverse effect is osteonecrosis of the jaw, frequently seen in oral and maxillofacial surgical patient care. After establishing a diagnosis, the treatment consists of surgical removal of necrotic bone while attempting to preserve viable bone. To date, suitable methods to accurately determine bone vitality remain deficient. Accordingly, patients are often confronted with recurrence of the disease, leading to subsequent further loss of jawbone with tremendous implications for ability of nutrition, speech, social interaction and overall quality of life. In this project, we introduce Raman spectroscopy to measure bone tissue viability, which is validated through high-resolution imaging methods. This merely optical, non-invasive technique is a promising candidate to ameliorate both diagnosis and treatment of patients affected by this debilitating disease.

    Contact and affiliation:

    Dr. med. Levi Matthies
    Department of Oral and Maxillofacial Surgery
    l.matthies@uke.de
    +49 (0) 40 7410 - 53259


  • Understanding the local tissue quality changes within human spinal metastases of breast and prostate cancer

    The underlying regulatory mechanisms of impaired bone matrix quality in metastatic spinal disease are poorly understood. Furthermore, its relation to metastatic growth, increasing the risk of paralysis due to spinal cord constriction (Figure 1) and fragility fractures, are not entirely elucidated yet. When cancer cells settle into bone, osteolytic or osteosclerotic lesions are caused by manipulating the activity of the bone-forming osteoblasts and bone-resorbing osteoclasts, at the same time benefiting from factors normally highly secreted by osteocytes i.e. sclerostin and RANKL via the metastatic cells. Bone stores growth factors and minerals that, when released, contribute to tumor progression. Bone matrix turnover is regulated by the mechanosensitive osteocyte network. This network of osteocytes might act as a central player, since I) osteocytes are regulators of bone remodeling, are II) actively participate in remodeling, and III) with their vastly active surface and interconnectivity could be the source of tumor cell-anabolic stimuli. We have designed a translational, interdisciplinary project utilizing a multitude of biological and material science techniques on precious human bone specimens of cancer patients. In this project we aim to assess the role of osteocytes in spinal bone metastases of breast and prostate cancer.

    Contact and affiliation:

    Dr. med. Annika Heuer
    Department Trauma and Orthopedic Surgery
    ann.heuer@uke.de

  • LiquidSWITCH – Early detection of resistance to BRAF inhibition and switch to immune checkpoint inhibition triggered by serial monitoring of cell-free DNA in blood plasma of melanoma patients.

    Signal transduction inhibition as well as Immune-checkpoint inhibition (ICI) have revolutionized the treatment of patients with metastatic melanoma. However, patients with metastatic melanoma harboring BRAF mutations are still first treated with targeted therapy (TT) consisting of combined BRAF- and MEK-inhibitors until acquired resistance develops, and then switched to ICI. A radiological assessment following RECIST 1.1. criteria is the current standard definition of resistance. Nevertheless, the subsequent switch to ICI can be too late to improve patient overall survival. Therefore, there is an unmet need to identify predictive biomarkers of early resistance to TT. The aim of this project is to establish a biobank und analyze ctDNA and circulating tumor cells (CTC) from patients with non-resectable metastatic melanoma via digital droplet PCR (ddPCR) and next generation sequencing (NGS) and to characterize them at the DNA and RNA level. Within the framework of a multicenter, observational study four collaborating skin cancer centers in Northern Germany (UKE Hamburg, UKSH Lübeck, UKSH Kiel, Elbekliniken Buxtehude) will participate in patient recruitment. The analyses will be performed at the ITB (UKE) in collaboration with the UKSH Lübeck. Proof-of-principle data from this study will provide the basis for follow on studies with sufficient power to assess whether tumors responding to BRAF inhibition as defined by response in ctDNA can improve efficacy of immune therapy and vice versa.

    Contact and affiliation:

    Dr. med. Isabel Heidrich
    Department of Dermatology/ Institute for Tumor Biology
    i.heidrich@uke.de
    +49 (0) 1522 - 2831042


  • Radiosensitization through molecular targeting in HNSCC tissue slice cultures of primary tumors and lymph node metastases

    Curative intended radiochemotherapy in human papillomavirus (HPV)-negative head and neck cancer, especially when presenting with lymph node metastasis, is associated with high rates of locoregional failure and tumor relapse. This demonstrates the necessity for more efficacious concepts. Benefits made in precision cancer medicine could complement radiotherapy but the interplay of targeted agents with radiation is mostly unknown. Considering HNSCC heterogeneity and the lack of predictive biomarkers, patient-derived short-term assay systems may close this gap. In this project, we assess the potential of HNSCC tissue slice cultures of primary and lymph node metastasis as patient-specific tumor models for pretherapeutic testing of potentially radiosensitizing agents. Due to previous work we focus on two promising strategies: 1. Inhibition of hyperactivated src family kinases (SFK) and 2. Targeting of the cellular DNA damage response (DDR) and DNA repair machinery through dual inhibition of PARP and Wee1.

    Contact and affiliation:

    Dr. med. Henrike Zech
    Department of Otorhinolaryngology
    +49 (0) 152 22824064
    h.zech@uke.de

  • Deciphering the Role of the Lymphotoxin – IL-22BP Axis in CRC metastasis

    We have recently described how IL-22BP (interleukin-22 binding protein), a natural antagonist of interleukin-22, bridges the antitumorigenic effects of lymphotoxin in colorectal cancer (CRC) in mice and humans (Kempski*, Giannou* et al., Gastroenterology, 2020). Furthermore, we have shown that IL-22 promotes the first steps of metastasis formation. Our hypothesis is that IL-22BP is upregulated at later stages and controls the progression of already established metastasis. Preliminary data indeed show a dysregulated expression of IL-22BP in both the primary tumor of patients with metastasized CRC and in the liver metastasis themselves. However, the cellular source and regulation of IL-22BP in liver metastasis remain unknown. Our preliminary data indicate that the major producer of IL-22BP in metastasis is a different DC subset compared to the primary tumor in the intestine. Our next aim is to examine whether lymphotoxin regulates IL-22BP expression in this DC subset. Finally, we want to study whether the pharmacological inhibition of lymphotoxin signaling or the dendritic cell-specific knock-out of RelB (part of the non-canonical NFkB signaling pathway activated by lymphotoxin) can influence liver metastasis progression and might thus represent a novel therapeutic target for the treatment of metastasized CRC.

    Contact and affiliation:

    Dr. med. Jan Kemspki
    Department of Internal Medicine I
    +49 152 22829943
    j.kempski@uke.de

  • Project 1: Intraventricular application of sonic-hedgehog pathway inhibition as a therapeutic option in young patients with medulloblastoma

    Medulloblastoma is the most common malignant brain tumor in children, arises in the posterior fossa, and disseminates via the cerebrospinal fluid. One of 4 molecular medulloblastoma subgroups is caused by Sonic Hedgehog (SHH) overactivation. The small molecule Vismodegib allosterically inhibits Smoothened (SMO), the main upstream activator of SHH. Unfortunately, Vismodegib has shown to cause severe bone growth defects in preclinical studies and a phase II clinical trial, preventing its systemic application in children. Therefore, we aimed to establish an intraventricular therapy with Vismodegib, combining the benefits of targeted drug delivery to the location of the tumor and metastases and minimizing systemic side effects. In a mouse model for SHH medulloblastoma, we compared intraventricular, oral, and placebo treatment regarding effects on survival, tumor biology and bone morphology. Our results show a significant survival benefit of intraventricularly treated mice compared to placebo and no signs of systemic side effects on bone growth. Our experimental work is almost concluded, and we will now prepare a manuscript for publication.

    Project 2: Molecular and clinical characterization of atypical peripheral nerve sheath tumors in NF1patients and prediction of risk for malignant transformation and dissemination

    Neurofibromatosis Type 1 (NF1) is a tumor predisposition syndrome that causes a broad spectrum of benign and malignant tumors. Most frequent are neurofibromas (NF)– benign tumors that are divided into several different subtypes and that are associated with a variety of clinical complications (Philpott et al. 2017). Atypical neurofibroma (ANF) are believed to be at risk for progression to highly aggressive malignant peripheral nerve sheath tumors (MPNST) that have a dismal prognosis and are considered incurable upon occurrence of metastasis (Korfhage und Lombard 2019). Therefore, the identification of such lesions is of high importance for risk adapted patient care and could help to reduce the mortality of NF1 patients. Based on small series, different histological and imaging criteria have been proposed to identify ANF or “atypical neurofibromatous neoplasms of uncertain biological potential” (ANNUBPs), but a consensus pathological definition has not yet been reached (Miettinen et al. 2017). Most importantly, a thorough molecular and clinical characterization is missing completely to date. In this project, we aim to characterize the molecular, histological and clinical attributes that distinguish ANF with high potential of malignant transformation and metastasis from benign NF and malignant MPNSTs. To this end, we will perform in-depth histological analysis as well as global DNA methylation, copy number profiling, and sequencing of 30 ANF and compare these data with existing reference cohorts of NF and MPNST. Finally, we will investigate clinical parameters and outcome of ANF patients, including the type of NF1 germline mutation, patients’ age, tumor localization, malignant tumor transformation, potential metastatic spread and long-term follow-up. Respective results shall serve as a solid base for a tailored surveillance and future clinical trials for patients with ANF.

    Contact and affiliation:

    Dr. med. Catena Kresbach
    Department of Neuropathology
    Research Institute Children's Cancer Center Hamburg
    +49 (40) 426051220
    c.kresbach@uke.de

  • Disentangling the pathomechanisms of arterial and venous thromboinflammation under immune checkpoint inhibitors

    Patients receiving ICI are at increased risk of developing venous thromboembolism (VTE) and cardiovascular adverse events. While large retrospective analyses confirm this observation, clinical studies on the underlying pathomechanisms are urgently needed to define potential therapeutic and preventive targets. My plan is to fill this knowledge gap by setting up a prospective clinical study on cancer patients undergoing treatment with ICI. I aim to investigate venous tissue factor (TF)-initiated coagulation activation and platelet and monocyte-driven atherosclerosis. By analyzing monocyte and microvesicle (MV)-TF activity, investigating platelet activity via platelet-platelet and platelet-monocyte aggregation studies, and implementing flow cytometry-based phenotyping of monocytes and platelets, I aim to elucidate the cellular drivers of ICI-mediated thromboinflammation in cancer patients.

    Contact and affiliation:

    Dr. med. Christina Rolling
    Department of Internal Medicine II
    c.rolling@uke.de

  • The Role of innate (NK) and adaptive (T) effector lymphocytes in themicroenvironment of soft-tissue sarcomas: a comprehensive investigation usinghuman-derived sarcoma-organoids

    Sarcomas are a group of heterogeneous malignancies rising from mesenchymal tissues. Despite theirrarity, a lack of sufficient systemic therapy strategies make sarcomas a challenging and clinically relevantsubject of basic scientific research6,7. While upcoming immunotherapies in different oncological fieldsshow promising results, up to this day, such approaches don’t play a relevant role in sarcoma-oncology8.With this project we would like to elucidate a new field of immunotherapy where it ironically got startedover a hundred years ago: in sarcomas9. To do so we aim to achieve a broader understanding of theimmunological microenvironment of sarcomas to investigate mechanisms which allow for immuneescape from both the innate and adaptive immune system. To study such mechanisms, we are currentlyanalyzing tumor infiltrating lymphocytes as well as cancer cells from patient-derived human sarcomasamples. We established patient-derived three-dimensional tumoroid models to evaluate humanlymphocyte-sarcoma interaction in vitro using the donors’ own tissue- and blood-derived lymphocytes.By expanding the understanding of lymphocyte-sarcoma interaction in those three-dimensional in vitromodels,we want to provide new approaches for immuno- and/or personalized sarcoma-therapy.

    Contact and affiliation:

    Dr. med. Adrian Sagebiel
    Klinik und Poliklinik für Allgemein- Viszeral- und Thoraxchirurgie
    a.sagebiel@uke.de

  • PIMPLE -Peripheral Immune Mediators in the Prognosis of diverse maLignanciEs

    The profound success of immune checkpoint inhibitors (ICI) in oncological therapy highlights the relevance of the immune system in malignancies. However, the role of soluble immune checkpoints (sICs) in antitumor immunity remains mostly unclear. In my own preliminary work a significant association between the levels of different sICs (sBTLA, sCD80, sTIM-3) in serum and overall survival of individuals with advanced solid malignancies before and during therapy with ICIs was shown. The levels of sICs in serum correlated with the expression of membrane-bound isoforms (ICs) on peripheral blood mononuclear cells (PBMCs). The aim of this project is to investigate sICs in an extended cohort including both hematologic and solid neoplasms with regard to their utility in predicting overall survival and treatment response. For this purpose, 14 different sICs will be determined in the plasma of these oncological patients using a multiplex immunoassay. Furthermore, the relationship between soluble and membrane-bound isoforms (ICs) on PBMCs and tumor-infiltrating lymphocytes (TILs), as well as their relationship to the microbiome, will be explored. A more detailed understanding of these easily measurable sICs could have diagnostic and prognostic, as well as therapeutic consequences in oncological diseases.

    Contact and affiliation:

    Dr. med. Joao Gorgulho
    Department of Medicine II Oncology, Hematology and Bone marrow transplantation with section Pneumology
    j.gorgulho@uke.de

  • Patient specific drug screening and biomarker research in pancreatic ductal adenocarcinoma using newly established tumoroids

    Partner: Dr. Tabea Sturmheit, 2cureX GmbH

    Pancreatic ductal adenocarcinoma (PDAC) has high metastatic potential and poorly efficient available therapies. Curative surgery is possible in 20% of patients and usually followed by adjuvant chemotherapeutic treatment, another approximately 20% with locally advanced non-metastastic disease may become candidates for secondary resection after neoadjuvant chemotherapy. Only 1/3 of patients respond to chemotherapy. The response cannot reliably be predicted due to lacking biomarkers]. A novel technology for drug response profiling is patient-derived three-dimensional (3D) tumor organoids (tumoroids). Since tumoroids show patient-specific features, they can be used as patient-specific avatars of disease and utilized to investigate drug response profiles. Establishing PDAC tumoroids has been proved challenging and only few European study groups were able to demonstrate initial positive results. Together with Dr. Tabea Sturmheit from 2cureX, a leading company for tumoroids and drug testing, the applicants’ group has started a collaboration to study the successful establishment of growing PDAC tumoroids - aiming to establish a reliable patient-derived PDAC tumoroid protocol, which will be used to establish individual chemosensitivity testing protocols. In a parallel liquid biopsy approach, patients’ mutation status will be assessed and correlated with individual tumoroid responses to different chemotherapeutic regimes.

    Contact and affiliation:

    Dr. med. Christine Sophia Nitschke
    Department of General, Visceral and Thoracic Surgery
    +49 (0) 152 22815806
    c.nitschke@uke.de

    Dr. Tabea Sturmheit
    2cureX GmbH
    +49 (0) 40 5259 4850
    tms@2cureX.com

  • Composition of the tumor microenvironment and immune repertoire in pediatric ALK-positive anaplastic large cell lymphoma

    Partners: Prof. Dr. Wolfram Klapper and Prof. Dr. Monika Brüggemann, UKSH Campus Kiel

    Anaplastic lymphoma kinase (ALK)-fusion protein-positive malignancies such as ALK-positive non-small cell lung cancer and ALK-positive anaplastic large cell lymphoma (ALCL) represent an ideal model forstudying tumor immunology: The ALK fusion protein is the driver of tumorigenesis, is almost exclusively expressed in the tumor cells and is recognized by the patient´s immune system. Pediatric ALK-positive ALCL patients mount an immune response against ALK characterized by the production of anti-ALK-antibodies and ALK-specific T-lymphocytes. The strength of this immune response is of high prognostic value. Little is known on the composition and competence of the immune cells in the microenvironment of pediatric ALCL. We propose a cooperative project together with the UKSH Kiel to (1) quantitatively describe the composition of the tumor microenvironment, (2) assess its functional capacity using gene expression analysis and (3) examine the T-cell receptor repertoire comparatively in blood and the microenvironment. Further elucidation of the host immune response against ALK-positive ALCL increases our understanding of the mechanisms behind the inter-individual strength of tumor-immunity. This forms the basis for the development of individualized therapeutic options like vaccination, check-pointinhibition or stem cell transplantation. Clinically, T cell receptor (TCR) repertoire analysis could represent a novel opportunity for long-term monitoring of patients.

    Contact and affiliation:

    Dr. med. Fabian Knörr
    Research Institute Children’s Cancer Center Hamburg and Department of Pediatric Hematology and Oncology
    f.knoerr@uke.de
    +49 (0) 40 7410 51219

    Prof. Dr. Monika Brüggemann
    Department of Internal Medicine II, UKSH, Campus Kiel
    Hämatologie Labor Kiel

    Prof. Dr. Wolfram Klapper
    Institute for Pathology, Section Hematopathology, UKSH, Campus Kiel

  • An integrated approach for drug target validation

    Partner: Prof. Dr. Matthias Wilmanns, EMBL Hamburg

    Distant metastasis is the major cause of cancer related death. Low RAI2 gene expression was initially identified at ITB/UCCH to be correlated with poor patient survival and in particular with the detection of disseminated tumor cells in the bone marrow of patients that were otherwise free of overt metastases. So far, the RAI2 protein function has not been annotated; but joint efforts of the applicant and the research team at EMBL funded by DFG have already shown that this protein acts as transcriptional co-regulator and that its inactivation in breast cancer cells directly leads and to dedifferentiation and deregulation of the estrogen response. This resulted in a high impact publication in Cancer Discovery (impact factor: 26.4). More recently both research teams showed that RAI2 counteracts oncogenic CtBP activity. Because active CtBPs suppress epithelial differentiation and propel cancer progression, we hypothesize that RAI2 mediated negative CtBP regulation might be exploited to treat cancer patients more efficiently. The major aim of the proposed project is to validate the potential of RAI2-mediated CtBP inactivation as new therapeutic concept using their integrated drug target validation approach and unique infrastructure at EMBL which will then become also available to other members of the MSNZ/UCCH consortium.

    Contact and affiliation:

    Dr. rer. nat. Stefan Werner
    Department of Tumor Biology
    st.werner@uke.de
    +49 (0) 40 7410 57947

    Prof. Dr. Matthias Wilmanns, PhD
    Head of EMBL (European Molecular Biology Laboratory) Hamburg
    EMBL Wilmanns Group