Startseite » Research Groups » Research Area 3: Targeted immunomodulatory therapy

Research Area 3: Targeted immunomodulatory therapy

Project H

Department of Internal Medicine V, Friedrich-Alexander Universität Erlangen-Nürnberg

Heads: Prof. Dr. Andreas Mackensen and Prof. Dr. Dimitrios Mougiakakos

Contact: andreas.mackensen(at)uk-erlangen.de , dimitrios.mougiakakos(at)uk-erlangen.de

Goals and research areas

  1. Optimization and development of T cell-based immunotherapies against cancer.
  2. Immunoregulatory cells in malignant diseases and stem cell transplantation
  3. Tumor metabolism and its impact on immune responses
  4. Development of innovative metabolic targeting in cancer

 

Methods:

Our groups employ a broad range of immunological, molecular and biochemical techniques. We have a strong translational focus and interconnection with clinics is very strong. Access to state-of-the-art equipment within brand-new laboratories is granted including amongst others 12-color flow cytometry, laser-scanning microscopy and metabolic flux analyzers.

 

Projects within i-Target

  • Project H: Harnessing immune responses in hematological malignancies by redox-remodeling

    Brief description: Emerging evidence suggest that tumor-associated oxidative stress weakens the patients’ immune system and thereby contributes to tumor immune escape and attenuates the efficacy of immune based therapies. In this PhD project we aim to (1) evaluate oxidative stress-induced immune alterations in patients with leukemia and to develop strategies for pharmacologically (2) antagonizing this metabolic condition and for (3) enhancing the resilience of immune cells towards it. The ultimate goal is to boost intrinsic anti-tumor immune responses and to increase the efficacy of immune based therapies. The methods utilized in this translational project will comprise a broad variety of cellular, immunological, and metabolic techniques.

 

Selected publications

  1. Jitschin R, Hofmann AD, Bruns H, Gießl A, Bricks J, Berger J, Saul D, Eckart MJ, Mackensen AMougiakakos D:
    Mitochondrial metabolism contributes to oxidative stress and reveals therapeutic targets in chronic lymphocytic leukemia:
    Blood 2014; 123 (17):2663-72.
  2. Aigner M, Feulner J, Schaffer S, Kischel R, Kufer P, Schneider K, Henn A, Rattel B, Friedrich M, Baeuerle PA, Mackensen A, Krause SW:
    T lymphocytes can be effectively recruited for ex vivo and in vivo lysis of AML blasts by a novel CD33/CD3-bispecific BiTE antibody construct.
    Leukemia 2013; 27:1107-15.
  3. Mougiakakos D, Jitschin R, von Bahr L, Poschke I, Gary R, Sundberg B, Gerbitz A, Ljungman P, Le Blanc K: 
    Immunosuppressive CD14+HLA-DRlow/neg IDO+ myeloid cells in patients following allogeneic hematopoietic stem cell transplantation.
    Leukemia 2013; 27 (2):377-88.
  4. Mougiakakos D, Jitschin R, Johansson CC, Okita R, Kiessling R, Le Blanc K: The impact of inflammatory licensing on the heme oxygenase-1 mediated induction of regulatory T-cells by human mesenchymal stem cells.
    Blood 2011; 117:4826-35.
  5. Fischer K, Voelkl S, Berger J, Andreesen R, Pomorski T, Mackensen A:
    Antigen recognition induces phosphatidylserine exposure on the cell surface of human CD8+ T cells. 
    Blood 2006; 108:4094-101.

 

Project I

Molecular Oncology Group, Department of Radiotherapy and Radiation Oncology, Klinikum der Universität München

Head: Prof. Dr. rer. nat. Kirsten Lauber

Contact: kirsten.lauber(at)med.uni-muenchen.de

Goals and research areas

  1. Molecular mechanisms of cell death and its modulation by targeted agents.
  2. Interaction of dying cells with the innate immune system and its utilization for the induction of adaptive anti-tumor immune responses.
  3. Cellular and molecular mechanisms, which contribute to the onset of inflammatory side effects in the context of radiotherapy, e.g. irradiation-induced pneumonitis.

Methods

We are using a wide spectrum of cell biological, molecular biological, and immunological methods. Different in vitro cell culture systems and in vivo tumor models are employed. Our group has access to state-of-the-art techniques and platforms, including multi-color flow cytometry, deconvolution flurorescence microscopy, and multimodal in vivo imaging.

 

Project within i-Target

  • Project I: Combination of radiotherapy with Hsp90 inhibition to enhance the extent and the immunogenicity of tumor cell death for the treatment of sarcomas.

Brief description: The induction of immunogenic forms of cell death by specific radiotherapeutic regimes appears to be a promising approach for the stimulation of innate and adaptive anti-tumor immune responses. The aim of this PhD project is to explore, whether Hsp90 inhibition in combination with radiotherapy can enhance the extent as well as the immunogenicity of sarcoma cell death, and whether this can contribute to the stimulation of anti-sarcoma immune responses. To this end, various molecular, cell biological, and immunological methods will be employed, including in vivo experiments.

 

Selected publications

  1. Stark K, Eckart A, Haidari S, Tirniceriu A, Lorenz M, von Bruhl ML, Gartner F, Khandoga AG, Legate KR, Pless R, Hepper I, Lauber K, Walzog B, Massberg S: Capillary and arteriolar pericytes attract innate leukocytes exiting through venules and 'instruct' them with pattern-recognition and motility programs. Nat Immunol 2013, 14(1):41-51.
  2. Orth M, Lauber K, Niyazi M, Friedl AA, Li M, Maihofer C, Schuttrumpf L, Ernst A, Niemoller OM, Belka C: Current concepts in clinical radiation oncology. Radiat Environ Biophys 2014, 53(1):1-29.
  3. Lauber K, Keppeler H, Munoz LE, Koppe U, Schroder K, Yamaguchi H, Kronke G, Uderhardt S, Wesselborg S, Belka C, Nagata S, Herrmann M: Milk fat globule-EGF factor 8 mediates the enhancement of apoptotic cell clearance by glucocorticoids. Cell Death Differ 2013, 20(9):1230-1240.
  4. Rosenwald M, Koppe U, Keppeler H, Sauer G, Hennel R, Ernst A, Blume KE, Peter C, Herrmann M, Belka C, Schulze-Osthoff K, Wesselborg S, Lauber K: Serum-derived plasminogen is activated by apoptotic cells and promotes their phagocytic clearance. J Immunol 2012, 189(12):5722-5728.
  5. Peter C, Waibel M, Keppeler H, Lehmann R, Xu G, Halama A, Adamski J, Schulze-Osthoff K, Wesselborg S, Lauber K: Release of lysophospholipid 'find-me' signals during apoptosis requires the ATP-binding cassette transporter A1. Autoimmunity 2012, 45(8):568-573.
  6. Lauber K, Ernst A, Orth M, Herrmann M, Belka C: Dying cell clearance and its impact on the outcome of tumor radiotherapy. Front Oncol 2012, 2:116.
  7. Blume KE, Soeroes S, Keppeler H, Stevanovic S, Kretschmer D, Rautenberg M, Wesselborg S, Lauber K: Cleavage of annexin A1 by ADAM10 during secondary necrosis generates a monocytic "find-me" signal. J Immunol 2012, 188(1):135-145.

 

Projects J and K

Molecular Immunology Group, Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich

Head: Prof. Dr. Jürgen Ruland

Contact: j.ruland@tum.de

 

Goals and research areas

Our research concentrates on the molecular mechanisms of immune cell activation and of the pathogenesis of cancer. By investigating following questions we want to provide targets for the therapeutic manipulation of the immune system:

  1. How do normal immune cells recognize pathogens and by which mechanisms does this recognition initiate immune defense activation?
  2. How do pathologically deregulated signals in blood cells lead to malignant transformation and thus to the development of leukemia or lymphoma?

 

Methods

We use a wide range of molecular, cellular, immunological and transgenic techniques with a focus on preclinical models. We are partner of the DKTK (German Consortium for Translational Cancer Research) and the German Center for Infection Research (DZIF). Additionally, we are well embedded in the Munich biomedical research community and amongst others have access to state-of-the-art array and sequencing technologies.

 

Projects within i-Target

  • Project J: Mechanisms of MALT1 in antigen receptor signalling

    Brief description: The protease MALT1 forms together with CARD11 and BCL10 a protein complex that is crucial for antigen receptor-mediated NF-κB signaling for lymphocyte activation and differentiation. We have genetically demonstrated that a selective inhibition of Malt1 protease function enhances inflammatory pathway by inhibiting regulatory T cells (Gewies et al., Cell Rep 2014). Nevertheless, the mechanisms of Malt1 function for lymphocyte regulation are still not well known. A wide array of molecular, cellular, immunological and transgenic techniques, including primary cell culture, flow cytometry and western blotting, will be used to elucidate the functional relevance of Malt1 in antigen receptor signaling.

  • Project K: Functional role of CARD11/BCL10/MALT1 complexes in lymphoid malignancy

    Brief description: Altered CBM signaling can be found in a number of lymphoid malignancies with hyperactivation of the CBM complex being a hallmark of lymphomagenesis. To mechanistically study the effects of putative interaction partners of CBM complex members on downstream signaling processes and to investigate their role in driving pathogenesis of lymphoid malignancies, different molecular, cellular and transgenic techniques will be used, including flow cytometry, primary patient sample analyses and inhibitor assays.

 

Selected Publications

  1. Wartewig T, Kurgyis Z, Keppler S, Pechloff K, Hameister E, Ollinger R, Maresch R, Buch T, Steiger K, Winter C, Rad R, Ruland J (2017). PD-1 is a haploinsufficient suppressor of T cell lymphomagenesis. Nature 552, 121-125.
  2. Roth S, Rottach A, Lotz-Havla A, Laux V, Muschaweckh A, Gersting SW, Muntau AC, Hopfner K-P, Jin L, Vanness K, Petrini JHJ, Drexler I, Leonhardt H, Ruland J (2014). Rad50 / Card9 interactions link cytosolic DNA sensing to IL-1β production. Nat Immunol. 15, 538-545.
  3. Neumann K, Castineiras-Vilarino M, Hockendorf U, Hannesschlager N, Lemeer S, Kupka D, Meyermann S, Lech M, Anders H J, Kuster B, Busch D H, Gewies A, Naumann R, Gross O and Ruland J (2014). Clec12a Is an Inhibitory Receptor for Uric Acid Crystals that Regulates Inflammation in Response to Cell Death. Immunity 40, 389-399.
  4. Strasser D, Neumann K, Bergmann H, Marakalala M J, Guler R, Rojowska A, Hopfner K-P, Brombacher F, Urlaub H, Baier G, Brown G D, Leitges M and Ruland J (2012). Syk kinase-coupled C-type lectin receptors engage protein kinase C-sigma to elicit Card9 adaptor-mediated innate immunity. Immunity 36, 32-42.
  5. Gross O, Poeck H, Bscheider M, Dostert C, Hannesschläger N, Endres S, Hartmann G, Tardivel A, Schweighoffer E, Tybulewicz V, Mocsai A, Tschopp J, Ruland J (2009). Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence. Nature 459, 433-6.

 

Projekt L

Cancer Vaccine Lab, Division of Clinical Pharmacology, Department of Internal Medicine IV, Klinikum der Universität München

Head: Prof. Dr. Max Schnurr and Dr. Lars König

Contact: max.schnurr(at)med.uni-muenchen.de

 

Goals and research areas

  1. Mechanisms of tumor-induced immune suppression in pancreatic cancer
  2. Tumor immunotherapy based on recognition of cytosolic RNA species by RIG-I-like helikases
  3. Development of bifunctional, immunostimulatory RNA molecules for immunotherapy of gastrointestinal cancers
  4. Evaluation of 3p-RNA therapy-suited combination compounds (Checkpoint inhibitors, immunogenic cell death inducers)
  5. Mechanim of 3p-RNA induced cell death and immunological conseqences

 

Methods

Our group uses a wide range of immunological and molecular biology-based techniques. We are specialized in the preclinical testing of new anti-tumor therapies in relevant in vitro and in vivo models (pancreatic cancer, liver cancer). Our group has access to state-of-the-art devices such as multicolor-flow cytometry, confocal microscopy, cell sorter, generation of CRISPR/CAS9-mediated knock out cells, proteomics and next generation sequencing.

 

  • Project within i-Target:

Project L: Immunosuppression in gastrointestinal cancers using 3p-RNA-based combination therapies

Brief description: Pancreatic cancer is characterized by a potent immunosuppressive network entertained by tumor and stroma cells in the tumor microenvironment. Type I interferons (IFN) play a critical role in anti-tumor immunity. Synthetic 3p-RNA molecules activating RIG-I, a cytosolic sensor for viral RNA, can be exploited to induce a therapeutic type I IFN response as well as an immunogenic form of tumor cell death leading to the priming of adaptive anti-tumor immunity. In addition, by adding specific RNA sequences into 3p-RNA molecules siRNA-mediated gene silencing of tumor promoting or immunosuppressive molecules can be combined with RIG-I signaling in a single molecule. We demonstrated efficacy of this approach in several tumor entities, including pancreatic cancer und hepatocellular cancer (HCC). RNAseq of 3p-RNA treated tumor cells revealed potential “escape” mechanisms as well as vulnerabilities, which can be exploited for combinatorial approaches. These include immune escape and metabolic pathways. In addition, robot-assisted drug screening will be employed for an unbiased search for combination therapies using a large compound library. Promising candidates will be tested for in vivo efficacy in relevant murine tumor models..

Selected Publications

  1. Bauer C, Kühnemuth B, Duewell P, Ormanns S, Gress T, Schnurr M.
    Prevailing over T cell exhaustion: New developments in the immunotherapy of pancreatic

    Cancer Lett. 2016 Oct 10;381(1):259-68.

  2. Duewell P, Beller E, Kirchleitner SV, Adunka T, Bourhis H, Siveke J, Mayr D, Kobold S, Endres S, Schnurr M.
    Targeted activation of melanoma differentiation-associated protein 5 (MDA5) for immunotherapy of pancreatic carcinoma.

    OncoImmunology.2015; 4(10).

  3. Meng G, Xia M, Xu C, Yuan D, Schnurr M, Wei J.
    Multifunctional antitumor molecule 5'-triphosphate siRNA combining glutaminase silencing and RIG-I activation.

    Int J Cancer. 2014;134(8):1958-71.

  4. Duewell P, Steger A, Lohr H, Bourhis H, Hoelz H, Kirchleitner SV, Stieg MR, Grassmann S, Kobold S, Siveke JT, Endres S, Schnurr M.
    RIG-I-like helicases induce immunogenic cell death of pancreatic cancer cells and sensitize tumors toward killing by CD8(+) T cells.

    Cell Death Differ. 2014 Dec;21(12):1825-37.

  5. Ellermeier J, Wei J, Duewell P, Hoves S, Stieg MR, Adunka T, Noerenberg D, Anders HJ, Mayr D, Poeck H, Hartmann G, Endres S, Schnurr M.
    Therapeutic efficacy of bifunctional siRNA combining TGF-β1 silencing with RIG-I activation in pancreatic cancer.

    Cancer Res. 2013;73(6):1709-20.