Network


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

Hotspot


Dive into the research topics where Elke Malenke is active.

Publication


Featured researches published by Elke Malenke.


Molecular Cancer Therapeutics | 2008

Ritonavir induces endoplasmic reticulum stress and sensitizes sarcoma cells toward bortezomib-induced apoptosis

Marianne Kraus; Elke Malenke; Jeannette Gogel; Holger Müller; Thomas Rückrich; Herman S. Overkleeft; Huib Ovaa; Ewa Koscielniak; J. T. Hartmann; Christoph Driessen

The biosynthesis of immunoglobulin leads to constitutive endoplasmic reticulum (ER) stress in myeloma cells, which activates the unfolded protein response (UPR). The UPR promotes protein folding by chaperones and increases proteasomal degradation of misfolded protein. Excessive ER stress induces apoptosis and represents a molecular basis for the bortezomib sensitivity of myeloma. Most solid malignancies such as sarcoma, by contrast, are poorly bortezomib sensitive and display low levels of ER stress. We hypothesized that pharmacologic induction of ER stress might sensitize malignancies to bortezomib treatment. We show that the HIV protease inhibitor ritonavir induces ER stress in bortezomib-resistant sarcoma cells. Ritonavir triggered the UPR, decreased the degradation of newly synthesized protein, but did not directly inhibit proteasomal active sites in the therapeutic dose range in contrast to bortezomib. Whereas neither bortezomib nor ritonavir monotherapy translated into significant apoptosis at therapeutic drug levels, the combination strongly increased the level of ER stress and activated PERK, IRE1, and ATF6, synergistically induced CHOP, JNK, caspase-4, and caspase-9, and resulted in >90% apoptosis. In summary, ritonavir increases the level of ER stress induced by bortezomib, which sensitizes bortezomib-resistant cells to bortezomib-induced apoptosis. Ritonavir may therefore be tested clinically to improve the sensitivity of solid malignancies toward bortezomib treatment. [Mol Cancer Ther 2008;7(7):1940–8]


Blood | 2013

Steady-state neutrophil homeostasis is dependent on TLR4/TRIF signaling

Stefanie Bugl; Stefan Wirths; Markus P. Radsak; Hansjörg Schild; Pamela Stein; Maya C. André; Martin R. Müller; Elke Malenke; Tina Wiesner; Melanie Märklin; Julia-Stefanie Frick; Rupert Handgretinger; Hans-Georg Rammensee; Lothar Kanz; Hans-Georg Kopp

UNLABELLED Polymorphonuclear neutrophil granulocytes (neutrophils) are tightly controlled by an incompletely understood homeostatic feedback loop adjusting the marrows supply to peripheral needs. Although it has long been known that marrow cellularity is inversely correlated with G-CSF levels, the mechanism linking peripheral clearance to production remains unknown. Herein, the feedback response to antibody induced neutropenia is characterized to consist of G-CSF–dependent shifts of marrow hematopoietic progenitor populations including expansion of the lin-/Sca-1/c-kit (LSK) and granulocyte macrophage progenitor (GMP) compartments at the expense of thrombopoietic and red cell precursors. Evidence is provided that positive feedback regulation is independent from commensal germs as well as T, B, and NK cells. However, in vivo feedback is impaired in TLR4-/- and TRIF-/-, but not MyD88-/- animals. In conclusion, steady-state neutrophil homeostasis is G-CSF–dependent and regulated through pattern-recognition receptors,thereby directly linking TLR-triggering to granulopoiesis. KEY POINTS Steady-state and emergency granulopoiesis are both dependent on TLR signaling.


Investigational New Drugs | 2005

Induction of apoptosis by flavopiridol unrelated to cell cycle arrest in germ cell tumour derived cell lines

Frank Mayer; Sandra Mueller; Elke Malenke; M.A. Kuczyk; J. T. Hartmann; Carsten Bokemeyer

SummaryBackground: Germ cell tumours (GCTs) are highly sensitive to cisplatin-based chemotherapy. The inability to arrest the cell cycle at the G1/S-check-point due to a lack of retinoblastoma gene product RB has been suggested as one potential explanation for this feature. Flavopiridol (FP), an inhibitor of cyclin dependent kinases, causes cell cycle arrest or apoptosis depending on the relation of the transcription factor E2F1 and RB.Methods: The effect of FP was evaluated in GCT-derived cell lines NT2, 2102 EP and NCCIT in comparison to cell lines derived from ovarian cancer (SKOV), breast cancer (MCF7), and cervical cancer (HeLa) using the MTT-assay. Cell cycle progression and induction of apoptosis were assessed by flow cytometry and immunoblot analysis of PARP-cleavage.Results: FP did not affect cell cycle progression and proliferation of GCT cell lines at sublethal doses. At higher concentrations, cell death occurred independent of cell cycle progression. The IC50 was approximately fivefold lower for the three GCT cell lines (60/60/70 nM) than for the other tumour cell lines tested (350/280/300 nM). Lethal doses in vitro were markedly lower than plasma concentrations of FP achieved in clinical studies. In vitro sensitivity to FP did not correlate with that to cisplatin. The cell lines NTera2 and NCCIT showed comparable responses to FP despite differing in their IC50 to cisplatin by factor 4. Flow cytometry and immunoblot for PARP indicated apoptotic cell death induced by FP. Synergism between either cisplatin or paclitaxel and FP was not observed. However, at low concentrations, cytotoxicity of FP and cisplatin appeared to be additive.Conclusion: These prelinical investigations suggest a significant antitumour activity of FP in GCT. GCT derived cell lines were far more responsive to FP than cell lines derived from other solid tumours. In contrast to other models, FP did not induce cell cycle arrest in the GCT-derived cell lines tested, possibly due to the known lack of RB-expression in GCTs. However, apoptosis was induced unrelated to cell cycle progression already at low concentrations. No cross resistance between FP and cisplatin was observed. A clinical trial evaluating the activity of FP in patients with cisplatin-refractory GCTs appears to be warranted.


Stem Cells Translational Medicine | 2013

Shared Cell Surface Marker Expression in Mesenchymal Stem Cells and Adult Sarcomas

Stefan Wirths; Elke Malenke; Torsten Kluba; Simone Rieger; Martin R. Müller; Sabine Schleicher; Claus Hann von Weyhern; Florian Nagl; Falko Fend; Wichard Vogel; Frank Mayer; Lothar Kanz; Hans-Jörg Bühring; Hans-Georg Kopp

Advanced adult soft‐tissue sarcomas (STSs) are rare tumors with a dismal prognosis and limited systemic treatment options. STSs may originate from mesenchymal stem cells (MSCs); the latter have mainly been isolated from adult bone marrow as plastic‐adherent cells with differentiation capacity into mesenchymal tissues. Recently, a panel of antibodies has been established that allows for the prospective isolation of primary MSCs with high selectivity. Similar to cancer stem cells in other malignancies, sarcoma stem cells may bear immunophenotypic similarity with the corresponding precursor, that is, MSCs. We therefore set out to establish the expression pattern of MSC markers in sarcoma cell lines and primary tumor samples by flow cytometry. In addition, fibroblasts from different sources were examined. The results document a significant amount of MSC markers shared by sarcoma cells. The expression pattern includes uniformly expressed markers, as well as MSC markers that only stained subpopulations of sarcoma cells. Expression of W5C5, W8B2 (tissue nonspecific alkaline phosphatase [TNAP]), CD344 (frizzled‐4), and CD271 marked subpopulations displaying increased proliferation potential. Moreover, CD271+ cells displayed in vitro doxorubicin resistance and an increased capacity to form spheres under serum‐free conditions. Interestingly, another set of antigens, including the bona fide progenitor cell markers CD117 and CD133, were not expressed. Comparative expression patterns of novel MSC markers in sarcoma cells, as well as fibroblasts and MSCs, are presented. Our data suggest a hierarchical cytoarchitecture of the most common adult type sarcomas and introduce W5C5, TNAP, CD344, and CD271 as potential sarcoma progenitor cell markers.


BMC Research Notes | 2014

Paraneoplastic granulocyte colony-stimulating factor secretion in soft tissue sarcoma mimicking myeloproliferative neoplasia: a case report

Christiane Dorn; Stefanie Bugl; Elke Malenke; Martin R. Müller; Katja Weisel; Ulrich Vogel; Marius Horger; Lothar Kanz; Hans-Georg Kopp

BackgroundWhile paraneoplastic leukocytosis is a common phenomenon in solid tumors, extreme elevations of white blood counts (WBC) in the range of more than 100,000/μl are uncommon in patients with non-hematologic malignancies. Leukocytosis with mature neutrophils due to a granulocyte colony-stimulating factor (G-CSF) producing tumor is only seen on rare occasions.Case presentationMassive neutrophil leukocytosis of approximately 100,000/μl was diagnosed in a 57-year-old Caucasian woman with metastatic undifferentiated endometrial sarcoma. A bone marrow trephine biopsy revealed massively increased granulopoiesis, but no evidence of monoclonal myeloproliferative disease. After the primary tumor had been resected, white blood count (WBC) plummeted and went back to nearly normal levels within one week. With progressive metastatic disease, granulocyte colony-stimulating factor (G-CSF) plasma levels were found to be increased by 10-fold. White blood count (WBC) strictly correlated with tumor burden and response to chemotherapy. In the final stage of treatment resistent disease, white blood count (WBC) approximated 300,000/μl.ConclusionWe report on a granulocyte colony-stimulating factor (G-CSF) secreting undifferentiated endometrial sarcoma, which was associated with extreme neutrophil counts. White blood count (WBC) were closely correlated with tumor burden and associated with an aggressive clinical course. We suggest that paraneoplastic neutrophilia represents a poor prognostic sign in soft tissue sarcoma. In patients with similar constellations, antitumor therapy must not be delayed.


International Journal of Oncology | 2006

Cell-cycle progression and response of germ cell tumors to cisplatin in vitro

Sandra Mueller; Marcus M. Schittenhelm; Friedemann Honecker; Elke Malenke; Kirsten Lauber; Sebastian Wesselborg; Joerg Thomas Hartmann; Carsten Bokemeyer; Frank Mayer


Journal of Clinical Oncology | 2010

Polyadenosine diphosphate-ribose polymerase (PARP) inhibition as a means of protecting the inner ear from cisplatin (CDDP)-mediated ototoxicity without affecting antitumor efficacy in vitro.

F. Mayer; M. Mueller; Elke Malenke; A. Schrattenholz; Hans-Georg Kopp; H. Loewenheim


Journal of Clinical Oncology | 2017

Expression of neurotrophins and their receptors in adult sarcomas.

Elke Malenke; Christiane Dorn; Stefan Wirths; Martin R Mueller; Lothar Kanz; Hans-Georg Kopp


Journal of Clinical Oncology | 2016

Detection of MSC-like cells in soft tissue sarcoma cell lines and primary tumors

Stefan Wirths; Elke Malenke; T. Wiesner; H. Buehring; J. T. Hartmann; Hans-Georg Kopp


Blood | 2011

Neutropenia Does Not Delay Lymphopoietic Regeneration in NODSCIDcγ−/− Mice

Stefanie Bugl; Stefan Wirths; R Müller Martin; Märklin Melanie; Tina Wiesner; Elke Malenke; Hans-Georg Kopp

Collaboration


Dive into the Elke Malenke's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lothar Kanz

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar

Frank Mayer

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tina Wiesner

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge