Network


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

Hotspot


Dive into the research topics where Georg Greiner is active.

Publication


Featured researches published by Georg Greiner.


Allergy | 2014

The KIT D816V allele burden predicts survival in patients with mastocytosis and correlates with the WHO type of the disease

Gregor Hoermann; Karoline V. Gleixner; Graziella E. Dinu; Michael Kundi; Georg Greiner; Friedrich Wimazal; Emir Hadzijusufovic; Gerlinde Mitterbauer; Christine Mannhalter; Peter Valent; Wolfgang R. Sperr

KIT D816V is present in a majority of patients with systemic mastocytosis (SM). We determined the KIT D816V allele burden by quantitative real‐time PCR in bone marrow and peripheral blood of 105 patients with mastocytosis. KIT D816V was detected in 92/105 patients (88%). Significant differences in the median allele burden were observed between disease subgroups: cutaneous mastocytosis (0.042%), indolent SM (0.285%), smoldering SM (5.991%), aggressive SM (9.346%), and SM with associated hematologic non‐mast cell lineage disease (3.761%) (P < 0.001). The KIT D816V burden also correlated with serum tryptase (R = 0.5, P < 0.005) but not with mast cell infiltration in bone marrow or mediator symptoms. Moreover, the allele burden was of prognostic significance regarding survival (P < 0.01). Patients responding to cytoreductive therapy showed a significant decrease in KIT D816V (P < 0.05). To conclude, the KIT D816V burden correlates with the variant of mastocytosis, predicts survival, and is a valuable follow‐up parameter in SM.


Clinical Cancer Research | 2016

Identification of CD25 as STAT5-Dependent Growth Regulator of Leukemic Stem Cells in Ph+ CML.

Irina Sadovnik; Andrea Hoelbl-Kovacic; Harald Herrmann; Gregor Eisenwort; Sabine Cerny-Reiterer; Wolfgang Warsch; Gregor Hoermann; Georg Greiner; Katharina Blatt; Barbara Peter; Gabriele Stefanzl; Daniela Berger; Martin Bilban; Susanne Herndlhofer; Heinz Sill; Wolfgang R. Sperr; Berthold Streubel; Christine Mannhalter; Tessa L. Holyoake; Veronika Sexl; Peter Valent

Purpose: In chronic myelogenous leukemia (CML), leukemic stem cells (LSC) represent a critical target of therapy. However, little is known about markers and targets expressed by LSCs. The aim of this project was to identify novel relevant markers of CML LSCs. Experimental Design: CML LSCs were examined by flow cytometry, qPCR, and various bioassays. In addition, we examined the multipotent CD25+ CML cell line KU812. Results: In contrast to normal hematopoietic stem cells, CD34+/CD38− CML LSCs expressed the IL-2 receptor alpha chain, IL-2RA (CD25). STAT5 was found to induce expression of CD25 in Lin−/Sca-1+/Kit+ stem cells in C57Bl/6 mice. Correspondingly, shRNA-induced STAT5 depletion resulted in decreased CD25 expression in KU812 cells. Moreover, the BCR/ABL1 inhibitors nilotinib and ponatinib were found to decrease STAT5 activity and CD25 expression in KU812 cells and primary CML LSCs. A CD25-targeting shRNA was found to augment proliferation of KU812 cells in vitro and their engraftment in vivo in NOD/SCID-IL-2Rγ−/− mice. In drug-screening experiments, the PI3K/mTOR blocker BEZ235 promoted the expression of STAT5 and CD25 in CML cells. Finally, we found that BEZ235 produces synergistic antineoplastic effects on CML cells when applied in combination with nilotinib or ponatinib. Conclusions: CD25 is a novel STAT5-dependent marker of CML LSCs and may be useful for LSC detection and LSC isolation in clinical practice and basic science. Moreover, CD25 serves as a growth regulator of CML LSCs, which may have biologic and clinical implications and may pave the way for the development of new more effective LSC-eradicating treatment strategies in CML. Clin Cancer Res; 22(8); 2051–61. ©2015 AACR.


The FASEB Journal | 2014

CD52 is a molecular target in advanced systemic mastocytosis

Gregor Hoermann; Katharina Blatt; Georg Greiner; Eva Maria Putz; Angelika Berger; Harald Herrmann; Sabine Cerny-Reiterer; Karoline V. Gleixner; Christoph Walz; Konrad Hoetzenecker; Leonhard Müllauer; Andreas Reiter; Karl Sotlar; Veronika Sexl; Peter Valent; Matthias Mayerhofer

Advanced systemic mastocytosis (SM) is an aggressive hematopoietic neoplasm with poor prognosis and short survival times. So far, no curative therapy is available for affected patients. We have identified the cell surface antigen CD52 (CAMPATH‐1) as a molecular target expressed abundantly on the surface of primary neoplastic mast cells (MCs) in patients with advanced SM. In contrast, neoplastic MCs of patients with indolent SM and normal MCs expressed only low levels or did not express CD52. To study the mechanisms of CD52 expression and the value of this antigen as a potential therapeutic target, we generated a human MC cell line, designated MCPV‐1, by lentiviral immortalization of cord blood‐derived MC progenitor cells. Functional studies revealed that activated RAS profoundly promotes surface expression of CD52. The CD52‐targeting antibody alemtuzumab induced cell death in CD52+ primary neoplastic MCs obtained from patients with SM as well as in MCPV‐1 cells. NSG mice xenotransplanted with MCPV‐1 cells survived significantly longer after treatment with alemtuzumab (median survival: 31 d untreated vs. 46 d treated; P=0.0012). We conclude that CD52 is a novel marker and potential therapeutic target in neoplastic MCs in patients with advanced SM.—Hoermann, G., Blatt, K., Greiner, G., Putz, E. M., Berger, A., Herrmann, H., Cerny‐Reiterer, S., Gleixner, K. V., Walz, C., Hoetzenecker, K., Müllauer, L., Reiter, A., Sotlar, K., Sexl, V., Valent, P., Mayerhofer, M. CD52 is a molecular target in advanced systemic mastocytosis. FASEB J. 28, 3540–3551 (2014). www.fasebj.org


Mediators of Inflammation | 2015

Cytokine Regulation of Microenvironmental Cells in Myeloproliferative Neoplasms

Gregor Hoermann; Georg Greiner; Peter Valent

The term myeloproliferative neoplasms (MPN) refers to a heterogeneous group of diseases including not only polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), but also chronic myeloid leukemia (CML), and systemic mastocytosis (SM). Despite the clinical and biological differences between these diseases, common pathophysiological mechanisms have been identified in MPN. First, aberrant tyrosine kinase signaling due to somatic mutations in certain driver genes is common to these MPN. Second, alterations of the bone marrow microenvironment are found in all MPN types and have been implicated in the pathogenesis of the diseases. Finally, elevated levels of proinflammatory and microenvironment-regulating cytokines are commonly found in all MPN-variants. In this paper, we review the effects of MPN-related oncogenes on cytokine expression and release and describe common as well as distinct pathogenetic mechanisms underlying microenvironmental changes in various MPN. Furthermore, targeting of the microenvironment in MPN is discussed. Such novel therapies may enhance the efficacy and may overcome resistance to established tyrosine kinase inhibitor treatment in these patients. Nevertheless, additional basic studies on the complex interplay of neoplastic and stromal cells are required in order to optimize targeting strategies and to translate these concepts into clinical application.


Journal of Leukocyte Biology | 2016

The tryptophan metabolite picolinic acid suppresses proliferation and metabolic activity of CD4+ T cells and inhibits c-Myc activation

Johanna Prodinger; Lisa J. Loacker; Ralf L. J. Schmidt; Franz Ratzinger; Georg Greiner; Nadine Witzeneder; Gregor Hoermann; Sabrina Jutz; Winfried F. Pickl; Peter Steinberger; Rodrig Marculescu; Klaus G. Schmetterer

Tryptophan metabolites, including kynurenine, 3‐hydroxyanthranilic acid, and picolinic acid, are key mediators of immunosuppression by cells expressing the tryptophan‐catabolizing enzyme indoleamine2,3‐dioxygenase. In this study, we assessed the influence of picolinic acid on cell viability and effector functions of CD4+ T cells following in vitro activation with agonistic anti‐CD3/anti‐CD28 antibodies. In contrast to kynurenine and 3‐hydroxyanthranilic acid, exposure of T cells with picolinic acid did not affect cell viability, whereas proliferation and metabolic activity were suppressed in a dose‐dependent manner. On the other hand, cytokine secretion and up‐regulation of cell surface activation markers were not or only weakly inhibited by picolinic acid. Picolinic acid exposure induced a state of deep anergy that could not be overcome by the addition of exogenous IL‐2 and inhibited Th cell polarization. On the molecular level, important upstream signaling molecules, such as the MAPKs ERK and p38 and the mammalian target of rapamycin target protein S6 ribosomal protein, were not affected by picolinic acid. Likewise, NFAT, NF‐κB, and AP‐1 promoter activity in Jurkat T cells was not influenced by exposure to picolinic acid. Whereas transcriptional levels of v‐myc avian myelocytomatosis viral oncogene homolog were not affected by picolinic acid, phosphorylation at Ser62 was strongly reduced in picolinic acid‐exposed T cells following activation. In conclusion, picolinic acid mediates a unique immunosuppressive program in T cells, mainly inhibiting cell cycle and metabolic activity, while leaving other effector functions intact. These functional features are accompanied by reduced phosphorylation of v‐myc avian myelocytomatosis viral oncogene homolog. It remains to be determined whether this effect is mediated by direct inhibition of ERK activity or whether indirect mechanisms apply.


Blood | 2017

CCL-2 is a KIT D816V-dependent Modulator of the Bone Marrow Microenvironment in Systemic Mastocytosis

Georg Greiner; Nadine Witzeneder; Angelika Berger; Klaus G. Schmetterer; Gregor Eisenwort; Ana-Iris Schiefer; Simone Roos; Theresia Popow-Kraupp; Leonhard Müllauer; Johannes Zuber; Veronika Sexl; Lukas Kenner; Wolfgang R. Sperr; Peter Valent; Matthias Mayerhofer; Gregor Hoermann

Systemic mastocytosis (SM) is characterized by abnormal accumulation of neoplastic mast cells harboring the activating KIT mutation D816V in the bone marrow and other internal organs. As found in other myeloproliferative neoplasms, increased production of profibrogenic and angiogenic cytokines and related alterations of the bone marrow microenvironment are commonly found in SM. However, little is known about mechanisms and effector molecules triggering fibrosis and angiogenesis in SM. Here we show that KIT D816V promotes expression of the proangiogenic cytokine CCL2 in neoplastic mast cells. Correspondingly, the KIT-targeting drug midostaurin and RNA interference-mediated knockdown of KIT reduced expression of CCL2. We also found that nuclear factor κB contributes to KIT-dependent upregulation of CCL2 in mast cells. In addition, CCL2 secreted by KIT D816V+ mast cells was found to promote the migration of human endothelial cells in vitro. Furthermore, knockdown of CCL2 in neoplastic mast cells resulted in reduced microvessel density and reduced tumor growth in vivo compared with CCL2-expressing cells. Finally, we measured CCL2 serum concentrations in patients with SM and found that CCL2 levels were significantly increased in mastocytosis patients compared with controls. CCL2 serum levels were higher in patients with advanced SM and were found to correlate with poor survival. In summary, we have identified CCL2 as a novel KIT D816V-dependent key regulator of vascular cell migration and angiogenesis in SM. CCL2 expression correlates with disease severity and prognosis. Whether CCL2 may serve as a therapeutic target in advanced SM remains to be determined in forthcoming studies.


Blood | 2018

Aggressive B-cell lymphomas in patients with myelofibrosis receiving JAK1/2 inhibitor therapy

Edit Porpaczy; Sabrina Tripolt; Andrea Hoelbl-Kovacic; Bettina Gisslinger; Zsuzsanna Bago-Horvath; Emilio Casanova-Hevia; Emmanuelle Clappier; Thomas Decker; Sabine Fajmann; Daniela A. Fux; Georg Greiner; Sinan Gueltekin; Gerwin Heller; Harald Herkner; Gregor Hoermann; Jean-Jacques Kiladjian; Thomas Kolbe; Christoph Kornauth; Maria-Theresa Krauth; Robert Kralovics; Leonhard Muellauer; Mathias Mueller; Michaela Prchal-Murphy; Eva Maria Putz; Emmanuel Raffoux; Ana-Iris Schiefer; Klaus G. Schmetterer; Christine Schneckenleithner; Ingrid Simonitsch-Klupp; Cathrin Skrabs

Inhibition of Janus-kinase 1/2 (JAK1/2) is a mainstay to treat myeloproliferative neoplasms (MPN). Sporadic observations reported the co-incidence of B-cell non-Hodgkin lymphomas during treatment of MPN with JAK1/2 inhibitors. We assessed 626 patients with MPN, including 69 with myelofibrosis receiving JAK1/2 inhibitors for lymphoma development. B-cell lymphomas evolved in 4 (5.8%) of 69 patients receiving JAK1/2 inhibition compared with 2 (0.36%) of 557 with conventional treatment (16-fold increased risk). A similar 15-fold increase was observed in an independent cohort of 929 patients with MPN. Considering primary myelofibrosis only (N = 216), 3 lymphomas were observed in 31 inhibitor-treated patients (9.7%) vs 1 (0.54%) of 185 control patients. Lymphomas were of aggressive B-cell type, extranodal, or leukemic with high MYC expression in the absence of JAK2 V617F or other MPN-associated mutations. Median time from initiation of inhibitor therapy to lymphoma diagnosis was 25 months. Clonal immunoglobulin gene rearrangements were already detected in the bone marrow during myelofibrosis in 16.3% of patients. Lymphomas occurring during JAK1/2 inhibitor treatment were preceded by a preexisting B-cell clone in all 3 patients tested. Sequencing verified clonal identity in 2 patients. The effects of JAK1/2 inhibition were mirrored in Stat1-/- mice: 16 of 24 mice developed a spontaneous myeloid hyperplasia with the concomitant presence of aberrant B cells. Transplantations of bone marrow from diseased mice unmasked the outgrowth of a malignant B-cell clone evolving into aggressive B-cell leukemia-lymphoma. We conclude that JAK/STAT1 pathway inhibition in myelofibrosis is associated with an elevated frequency of aggressive B-cell lymphomas. Detection of a preexisting B-cell clone may identify individuals at risk.


Scientific Reports | 2017

Chloroquine inhibits human CD4+ T-cell activation by AP-1 signaling modulation

Ralf L. J. Schmidt; Sabrina Jutz; Katrin Goldhahn; Nadine Witzeneder; Marlene Gerner; Doris Trapin; Georg Greiner; Gregor Hoermann; Guenter Steiner; Winfried F. Pickl; Heinz Burgmann; Peter Steinberger; Franz Ratzinger; Klaus G. Schmetterer

Chloroquine (CQ) is widely used as an anti-inflammatory therapeutic for rheumatic diseases. Although its modes of action on the innate immune system are well described, there is still insufficient knowledge about its direct effects on the adaptive immune system. Thus, we evaluated the influence of CQ on activation parameters of human CD4+ T-cells. CQ directly suppressed proliferation, metabolic activity and cytokine secretion of T-cells following anti-CD3/anti-CD28 activation. In contrast, CQ showed no effect on up-regulation of T-cell activation markers. CQ inhibited activation of all T helper cell subsets, although IL-4 and IL-13 secretion by Th2 cells were less influenced compared to other Th-specific cytokines. Up to 10 μM, CQ did not reduce cell viability, suggesting specific suppressive effects on T-cells. These properties of CQ were fully reversible in re-stimulation experiments. Analyses of intracellular signaling showed that CQ specifically inhibited autophagic flux and additionally activation of AP-1 by reducing phosphorylation of c-JUN. This effect was mediated by inhibition of JNK catalytic activity. In summary, we characterized selective and reversible immunomodulatory effects of CQ on human CD4+ T-cells. These findings provide new insights into the biological actions of JNK/AP-1 signaling in T-cells and may help to expand the therapeutic spectrum of CQ.


Haematologica | 2017

Combined targeting of STAT3 and STAT5: A novel approach to overcome drug resistance in chronic myeloid leukemia

Karoline V. Gleixner; Mathias Schneeweiss; Gregor Eisenwort; Daniela Berger; Harald Herrmann; Katharina Blatt; Georg Greiner; Konstantin Byrgazov; Gregor Hoermann; Marina Konopleva; Islam Waliul; Abbarna A. Cumaraswamy; Patrick T. Gunning; Hiroshi Maeda; Richard Moriggl; Michael W. Deininger; Thomas Lion; Michael Andreeff; Peter Valent

In chronic myeloid leukemia, resistance against BCR-ABL1 tyrosine kinase inhibitors can develop because of BCR-ABL1 mutations, activation of additional pro-oncogenic pathways, and stem cell resistance. Drug combinations covering a broad range of targets may overcome resistance. CDDO-Me (bardoxolone methyl) is a drug that inhibits the survival of leukemic cells by targeting different pro-survival molecules, including STAT3. We found that CDDO-Me inhibits proliferation and survival of tyrosine kinase inhibitor-resistant BCR-ABL1+ cell lines and primary leukemic cells, including cells harboring BCR-ABL1T315I or T315I+ compound mutations. Furthermore, CDDO-Me was found to block growth and survival of CD34+/CD38− leukemic stem cells (LSC). Moreover, CDDO-Me was found to produce synergistic growth-inhibitory effects when combined with BCR-ABL1 tyrosine kinase inhibitors. These drug-combinations were found to block multiple signaling cascades and molecules, including STAT3 and STAT5. Furthermore, combined targeting of STAT3 and STAT5 by shRNA and STAT5-targeting drugs also resulted in synergistic growth-inhibition, pointing to a new efficient concept of combinatorial STAT3 and STAT5 inhibition. However, CDDO-Me was also found to increase the expression of heme-oxygenase-1, a heat-shock-protein that triggers drug resistance and cell survival. We therefore combined CDDO-Me with the heme-oxygenase-1 inhibitor SMA-ZnPP, which also resulted in synergistic growth-inhibitory effects. Moreover, SMA-ZnPP was found to sensitize BCR-ABL1+ cells against the combination ‘CDDO-Me+ tyrosine kinase inhibitor’. Together, combined targeting of STAT3, STAT5, and heme-oxygenase-1 overcomes resistance in BCR-ABL1+ cells, including stem cells and highly resistant sub-clones expressing BCR-ABL1T315I or T315I-compound mutations. Whether such drug-combinations are effective in tyrosine kinase inhibitor-resistant patients with chronic myeloid leukemia remains to be elucidated.


British Journal of Haematology | 2017

Expansion of BCR/ABL1 + cells requires PAK2 but not PAK1

Leo Edlinger; Angelika Berger-Becvar; Ingeborg Menzl; Gregor Hoermann; Georg Greiner; Eva Grundschober; Zsuzsanna Bago-Horvath; Wael Al-Zoughbi; Gerald Hoefler; Christine Brostjan; Lars Gille; Richard Moriggl; Andreas Spittler; Veronika Sexl; Andrea Hoelbl-Kovacic

The p21‐activated kinases (PAKs) are key nodes in oncogenic signalling pathways controlling growth, survival, and motility of cancer cells. Their activity is increased in many human cancers and is associated with poor prognosis. To date, PAK deregulation has mainly been studied in solid tumours, where PAK1 and PAK4 are the main isoforms deregulated. We show that PAK1 and PAK2 are the critical isoforms in a BCR/ABL1+ haematopoietic malignancy. In suspension, leukaemic cells deficient for PAK1 and PAK2 undergo apoptosis, while the loss of either protein is well tolerated. Transfer of medium conditioned by shPAK2‐ but not shPAK1‐expressing leukaemic cells interferes with endothelial cell growth. We found that leukaemic cells produce exosomes containing PAK2. Transfer of isolated exosomes supports endothelial cell proliferation. In parallel, we found that leukaemic cells explicitly require PAK2 to grow towards an extracellular matrix. PAK2‐deficient cells fail to form colonies in methylcellulose and to induce lymphomas in vivo. PAK2 might therefore be the critical isoform in leukaemic cells by controlling tumour growth in a dual manner: vascularization via exosome‐mediated transfer to endothelial cells and remodelling of the extracellular matrix. This finding suggests that the PAK2 isoform represents a promising target for the treatment of haematological diseases.

Collaboration


Dive into the Georg Greiner's collaboration.

Top Co-Authors

Avatar

Gregor Hoermann

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Peter Valent

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Wolfgang R. Sperr

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Gregor Eisenwort

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Karoline V. Gleixner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Klaus G. Schmetterer

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Nadine Witzeneder

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Daniela Berger

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Katharina Blatt

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Matthias Mayerhofer

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge