Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maja Milanovic is active.

Publication


Featured researches published by Maja Milanovic.


Nature | 2013

Synthetic lethal metabolic targeting of cellular senescence in cancer therapy

Jan R. Dörr; Yong Yu; Maja Milanovic; Gregor Beuster; Christin Zasada; J. Henry M. Däbritz; Jan Lisec; Dido Lenze; Anne Gerhardt; Katharina Schleicher; Susanne Kratzat; Bettina Purfürst; Stefan Walenta; Wolfgang Mueller-Klieser; Markus Gräler; Michael Hummel; Ulrich Keller; Andreas K. Buck; Bernd Dörken; Lothar Willmitzer; Maurice Reimann; Stefan Kempa; Soyoung Lee; Clemens A. Schmitt

Activated oncogenes and anticancer chemotherapy induce cellular senescence, a terminal growth arrest of viable cells characterized by S-phase entry-blocking histone 3 lysine 9 trimethylation (H3K9me3). Although therapy-induced senescence (TIS) improves long-term outcomes, potentially harmful properties of senescent tumour cells make their quantitative elimination a therapeutic priority. Here we use the Eµ-myc transgenic mouse lymphoma model in which TIS depends on the H3K9 histone methyltransferase Suv39h1 to show the mechanism and therapeutic exploitation of senescence-related metabolic reprogramming in vitro and in vivo. After senescence-inducing chemotherapy, TIS-competent lymphomas but not TIS-incompetent Suv39h1– lymphomas show increased glucose utilization and much higher ATP production. We demonstrate that this is linked to massive proteotoxic stress, which is a consequence of the senescence-associated secretory phenotype (SASP) described previously. SASP-producing TIS cells exhibited endoplasmic reticulum stress, an unfolded protein response (UPR), and increased ubiquitination, thereby targeting toxic proteins for autophagy in an acutely energy-consuming fashion. Accordingly, TIS lymphomas, unlike senescence models that lack a strong SASP response, were more sensitive to blocking glucose utilization or autophagy, which led to their selective elimination through caspase-12- and caspase-3-mediated endoplasmic-reticulum-related apoptosis. Consequently, pharmacological targeting of these metabolic demands on TIS induction in vivo prompted tumour regression and improved treatment outcomes further. These findings unveil the hypercatabolic nature of TIS that is therapeutically exploitable by synthetic lethal metabolic targeting.


Genes & Development | 2011

Opposing roles of NF-κB in anti-cancer treatment outcome unveiled by cross-species investigations

Hua Jing; Julia Kase; Jan R. Dörr; Maja Milanovic; Dido Lenze; Michael Grau; Gregor Beuster; Sujuan Ji; Maurice Reimann; Peter Lenz; Michael Hummel; Bernd Dörken; Georg Lenz; Claus Scheidereit; Clemens A. Schmitt; Soyoung Lee

In malignancies, enhanced nuclear factor-κB (NF-κB) activity is largely viewed as an oncogenic property that also confers resistance to chemotherapy. Recently, NF-κB has been postulated to participate in a senescence-associated and possibly senescence-reinforcing cytokine response, thereby suggesting a tumor-restraining role for NF-κB. Using a mouse lymphoma model and analyzing transcriptome and clinical data from lymphoma patients, we show here that therapy-induced senescence presents with and depends on active NF-κB signaling, whereas NF-κB simultaneously promotes resistance to apoptosis. Further characterization and genetic engineering of primary mouse lymphomas according to distinct NF-κB-related oncogenic networks reminiscent of diffuse large B-cell lymphoma (DLBCL) subtypes guided us to identify Bcl2-overexpressing germinal center B-cell-like (GCB) DLBCL as a clinically relevant subgroup with significantly superior outcome when NF-κB is hyperactive. Our data illustrate the power of cross-species investigations to functionally test genetic mechanisms in transgenic mouse tumors that recapitulate distinct features of the corresponding human entity, and to ultimately use the mouse model-derived genetic information to redefine novel, clinically relevant patient subcohorts.


Nature | 2017

Senescence-associated reprogramming promotes cancer stemness

Maja Milanovic; Dorothy N.Y. Fan; Dimitri Belenki; J. Henry M. Däbritz; Zhen Zhao; Yong Yu; Jan R. Dörr; Lora Dimitrova; Dido Lenze; Inês Barbosa; Marco Antonio Mendoza-Parra; Tamara Kanashova; Marlen Metzner; Katharina Pardon; Maurice Reimann; Andreas Trumpp; Bernd Dörken; Johannes Zuber; Hinrich Gronemeyer; Michael Hummel; Gunnar Dittmar; Soyoung Lee; Clemens A. Schmitt

Cellular senescence is a stress-responsive cell-cycle arrest program that terminates the further expansion of (pre-)malignant cells. Key signalling components of the senescence machinery, such as p16INK4a, p21CIP1 and p53, as well as trimethylation of lysine 9 at histone H3 (H3K9me3), also operate as critical regulators of stem-cell functions (which are collectively termed ‘stemness’). In cancer cells, a gain of stemness may have profound implications for tumour aggressiveness and clinical outcome. Here we investigated whether chemotherapy-induced senescence could change stem-cell-related properties of malignant cells. Gene expression and functional analyses comparing senescent and non-senescent B-cell lymphomas from Eμ-Myc transgenic mice revealed substantial upregulation of an adult tissue stem-cell signature, activated Wnt signalling, and distinct stem-cell markers in senescence. Using genetically switchable models of senescence targeting H3K9me3 or p53 to mimic spontaneous escape from the arrested condition, we found that cells released from senescence re-entered the cell cycle with strongly enhanced and Wnt-dependent clonogenic growth potential compared to virtually identical populations that had been equally exposed to chemotherapy but had never been senescent. In vivo, these previously senescent cells presented with a much higher tumour initiation potential. Notably, the temporary enforcement of senescence in p53-regulatable models of acute lymphoblastic leukaemia and acute myeloid leukaemia was found to reprogram non-stem bulk leukaemia cells into self-renewing, leukaemia-initiating stem cells. Our data, which are further supported by consistent results in human cancer cell lines and primary samples of human haematological malignancies, reveal that senescence-associated stemness is an unexpected, cell-autonomous feature that exerts its detrimental, highly aggressive growth potential upon escape from cell-cycle blockade, and is enriched in relapse tumours. These findings have profound implications for cancer therapy, and provide new mechanistic insights into the plasticity of cancer cells.


Blood | 2017

Pharmacological restoration and therapeutic targeting of the B-cell phenotype in classical Hodgkin lymphoma

Jing Du; Martin Neuenschwander; Yong Yu; J. Henry M. Däbritz; Nina-Rosa Neuendorff; Kolja Schleich; Aitomi Bittner; Maja Milanovic; Gregor Beuster; Silke Radetzki; Edgar Specker; Maurice Reimann; Frank Rosenbauer; Stephan Mathas; Philipp Lohneis; Michael Hummel; Bernd Dörken; Jens Peter von Kries; Soyoung Lee; Clemens A. Schmitt

Classical Hodgkin lymphoma (cHL), although originating from B cells, is characterized by the virtual lack of gene products whose expression constitutes the B-cell phenotype. Epigenetic repression of B-cell-specific genes via promoter hypermethylation and histone deacetylation as well as compromised expression of B-cell-committed transcription factors were previously reported to contribute to the lost B-cell phenotype in cHL. Restoring the B-cell phenotype may not only correct a central malignant property, but it may also render cHL susceptible to clinically established antibody therapies targeting B-cell surface receptors or small compounds interfering with B-cell receptor signaling. We conducted a high-throughput pharmacological screening based on >28 000 compounds in cHL cell lines carrying a CD19 reporter to identify drugs that promote reexpression of the B-cell phenotype. Three chemicals were retrieved that robustly enhanced CD19 transcription. Subsequent chromatin immunoprecipitation-based analyses indicated that action of 2 of these compounds was associated with lowered levels of the transcriptionally repressive lysine 9-trimethylated histone H3 mark at the CD19 promoter. Moreover, the antileukemia agents all-trans retinoic acid and arsenic trioxide (ATO) were found to reconstitute the silenced B-cell transcriptional program and reduce viability of cHL cell lines. When applied in combination with a screening-identified chemical, ATO evoked reexpression of the CD20 antigen, which could be further therapeutically exploited by enabling CD20 antibody-mediated apoptosis of cHL cells. Furthermore, restoration of the B-cell phenotype also rendered cHL cells susceptible to the B-cell non-Hodgkin lymphoma-tailored small-compound inhibitors ibrutinib and idelalisib. In essence, we report here a conceptually novel, redifferentiation-based treatment strategy for cHL.


Molecular Cancer Therapeutics | 2016

CD20-Targeting Immunotherapy Promotes Cellular Senescence in B-Cell Lymphoma

J. Henry M. Däbritz; Yong Yu; Maja Milanovic; Martin Schönlein; Mathias T. Rosenfeldt; Jan R. Dörr; Andreas M. Kaufmann; Bernd Dörken; Clemens A. Schmitt

The CD20-targeting monoclonal antibody rituximab is an established component of immunochemotherapeutic regimens against B-cell lymphomas, where its coadministration with conventional anticancer agents has significantly improved long-term outcome. However, the cellular mechanisms by which rituximab exerts its antilymphoma activity are only partially understood. We show here that rituximab induces typical features of cellular senescence, a long-term growth arrest of viable cells with distinct biologic properties, in established B-cell lymphoma cell lines as well as primary transformed B cells. In addition, rituximab-based immunotherapy sensitized lymphoma cells to senescence induction by the chemotherapeutic compound adriamycin (a.k.a. doxorubicin), and, to a lesser extent, by the antimicrotubule agent vincristine. Anti-CD20 treatment further enhanced secretion of senescence-associated cytokines, and augmented the DNA damage response signaling cascade triggered by adriamycin. As the underlying prosenescence mechanism, we found intracellular reactive oxygen species (ROS) levels to be elevated in response to rituximab, and, in turn, the ROS scavenger N-acetylcysteine to largely abrogate rituximab-mediated senescence. Our results, further supported by gene set enrichment analyses in a clinical data set of chronic lymphocytic leukemia patient samples exposed to a rituximab-containing treatment regimen, provide important mechanistic insights into the biologic complexity of anti-CD20-evoked tumor responses, and unveil cellular senescence as a hitherto unrecognized effector principle of the antibody component in lymphoma immunochemotherapy. Mol Cancer Ther; 15(5); 1074–81. ©2016 AACR.


Trends in Cell Biology | 2018

The Senescence–Stemness Alliance – A Cancer-Hijacked Regeneration Principle

Maja Milanovic; Yong Yu; Clemens A. Schmitt

Activated oncogenes or anticancer therapies evoke senescent cell-cycle arrest in (pre-)malignant cells, thereby interrupting tumor formation or progression. Physiologically, cellular senescence contributes to embryonic development and tissue regeneration. These observations and the overlap of numerous gene products in senescence and stem cell signaling prompted investigations into whether epigenetic establishment of the senescent state may concomitantly reprogram the cell into a latent stem-like condition, whose functional impact becomes evident when arrested cells resume proliferation. We review here recent discoveries underscoring the unexpected senescence-stemness alliance, elucidate underlying molecular mechanisms, and discuss its fundamentally different implications in normal tissue repair - to replenish the exhausted repopulation capacity - as compared to cancer biology, where usurpation of this natural principle accounts for particularly aggressive tumor behavior.


Blood | 2014

Chemotherapy-Induced Senescence Reprograms Lymphoma and Leukemia Cells into Latent Cancer Stem Cells That Are Susceptible to Conceptually Novel Treatments

Clemens A. Schmitt; Maja Milanovic; Henry Daebritz; Zhen Zhao; Andreas Trumpp


Blood | 2013

Acquired Stem Cell Properties In Therapy-Induced Senescence Of Lymphomas and Acute Leukemias In Vitro and In Vivo

Maja Milanovic; Zhen Zhao; Jan R. Dörr; Yong Yu; Dido Lenze; Bernd Dörken; Michael Hummel; Christoph Dieterich; Scott W. Lowe; Clemens A. Schmitt


Blood | 2014

Elevated ROS Levels in Response to CD20-Targeting Enhance Senescence Entry after Immunochemotherapy in Human B-Cell Lymphoma

J. Henry M. Däbritz; Yong Yu; Maja Milanovic; Jan R. Dörr; Andreas M. Kaufmann; Mathias T. Rosenfeldt; Bernd Dörken; Clemens A. Schmitt


Blood | 2013

H3K9me3-Governed Senescence In Tumor Development and Lymphoma Treatment

Jan Dörr; Yong Yu; Maja Milanovic; Sujuan Ji; Sandra Spiesicke-Wegener; Philipp Lohneis; Bernd Dörken; Soyoung Lee; Clemens A. Schmitt

Collaboration


Dive into the Maja Milanovic's collaboration.

Top Co-Authors

Avatar

Yong Yu

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge