Network


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

Hotspot


Dive into the research topics where Christian Sillaber is active.

Publication


Featured researches published by Christian Sillaber.


American Journal of Hematology | 2011

Progressive peripheral arterial occlusive disease and other vascular events during nilotinib therapy in CML

Karl J. Aichberger; Susanne Herndlhofer; Gerit-Holger Schernthaner; Martin Schillinger; Gerlinde Mitterbauer-Hohendanner; Christian Sillaber; Peter Valent

The second generation BCR/ABL kinase inhibitor nilotinib is increasingly used for the treatment of imatinib‐resistant chronic myeloid leukemia (CML). So far, nilotinib is considered a well‐tolerated drug with little if any side effects, although an increase in the fasting glucose level has been reported. We examined a series of 24 consecutive CML patients treated with nilotinib in our center for the development of non‐hematologic adverse events. Three of these 24 CML patients developed a rapidly progressive peripheral arterial occlusive disease (PAOD) during treatment with nilotinib. In all three cases, PAOD required repeated angioplasty and/or multiple surgeries within a few months. No PAOD was known before nilotinib‐therapy in these patients, although all three had received imatinib. In two patients, pre‐existing risk factors predisposing for PAOD were known, and one of them had developed diabetes mellitus during nilotinib. In the other 21 patients treated with nilotinib in our center, one less severe PAOD, one myocardial infarction, one spinal infarction, one subdural hematoma, and one sudden death of unknown etiology were recorded. In summary, treatment with nilotinib may be associated with an increased risk of vascular adverse events, including PAOD development. In a subgroup of patients, these events are severe or even life‐threatening. Although the exact mechanisms remain unknown, we recommend screening for pre‐existing PAOD and for vascular risk factors such as diabetes mellitus in all patients before starting nilotinib and in the follow up during nilotinib‐therapy. Am. J. Hematol. 2011.


Blood | 2011

High STAT5 levels mediate imatinib resistance and indicate disease progression in chronic myeloid leukemia

Wolfgang Warsch; Karoline Kollmann; Eva Eckelhart; Sabine Fajmann; Sabine Cerny-Reiterer; Andrea Hölbl; Karoline V. Gleixner; Michael Dworzak; Matthias Mayerhofer; Gregor Hoermann; Harald Herrmann; Christian Sillaber; Gerda Egger; Peter Valent; Richard Moriggl; Veronika Sexl

In BCR-ABL1(+) leukemia, drug resistance is often associated with up-regulation of BCR-ABL1 or multidrug transporters as well as BCR-ABL1 mutations. Here we show that the expression level of the transcription factor STAT5 is another parameter that determines the sensitivity of BCR-ABL1(+) cells against tyrosine kinase inhibitors (TKIs), such as imatinib, nilotinib, or dasatinib. Abelson-transformed cells, expressing high levels of STAT5, were found to be significantly less sensitive to TKI-induced apoptosis in vitro and in vivo but not to other cytotoxic drugs, such as hydroxyurea, interferon-β, or Aca-dC. The STAT5-mediated protection requires tyrosine phosphorylation of STAT5 independent of JAK2 and transcriptional activity. In support of this concept, under imatinib treatment and with disease progression, STAT5 mRNA and protein levels increased in patients with Ph(+) chronic myeloid leukemia. Based on our data, we propose a model in which disease progression in BCR-ABL1(+) leukemia leads to up-regulated STAT5 expression. This may be in part the result of clonal selection of cells with high STAT5 levels. STAT5 then accounts for the resistance against TKIs, thereby explaining the dose escalation frequently required in patients reaching accelerated phase. It also suggests that STAT5 may serve as an attractive target to overcome imatinib resistance in BCR-ABL1(+) leukemia.


Cancer Research | 2004

Identification of Heme Oxygenase-1 As a Novel BCR/ABL-Dependent Survival Factor in Chronic Myeloid Leukemia

Matthias Mayerhofer; Stefan Florian; Maria-Theresa Krauth; Karl J. Aichberger; Martin Bilban; Rodrig Marculescu; Dieter Printz; Gerhard Fritsch; Oswald Wagner; Edgar Selzer; Wolfgang R. Sperr; Peter Valent; Christian Sillaber

Chronic myeloid leukemia (CML) is a stem cell disease in which BCR/ABL promotes the survival of leukemic cells. Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the degradation of heme and has recently been implicated in the regulation of growth and survival of various neoplastic cells. In the present study, we analyzed the expression and role of HO-1 in CML cells. As assessed by Northern and Western blot analysis as well as immunostaining, primary CML cells were found to express HO-1 mRNA and the HO-1 protein in a constitutive manner. Exposure of these cells to the BCR/ABL tyrosine kinase inhibitor STI571 resulted in decreased expression of HO-1 mRNA and protein. In addition, BCR/ABL was found to up-regulate HO-1 promoter activity, mRNA levels, and protein levels in Ba/F3 cells. To investigate the role of HO-1 for survival of primary CML cells, the HO-1 inducer hemin was used. Hemin-induced expression of HO-1 was found to protect CML cells from STI571-induced cell death. In addition, inhibition of HO-1 by zinc-(II)-deuteroporphyrin-IX-2,4-bisethyleneglycol resulted in a substantial decrease of cell viability. Furthermore, overexpression of HO-1 in the CML-derived cell line K562 was found to counteract STI571-induced apoptosis. Together, our data identify HO-1 as a novel BCR/ABL-driven survival molecule and potential target in leukemic cells in patients with CML. The pathogenetic and clinical implications of this observation remain to be elucidated.


Leukemia | 1997

Prognostic significance of WT1 gene expression at diagnosis in adult de novo acute myeloid leukemia

Schmid D; Heinze G; Linnerth B; Tisljar K; R Kusec; Klaus Geissler; Christian Sillaber; Klaus Laczika; Mitterbauer M; S Zöchbauer; Christine Mannhalter; Oskar A. Haas; Klaus Lechner; Ulrich Jäger; Alexander Gaiger

We examined the presence of WT1-specific mRNA in bone marrow samples of 125 patients with de novo acute myeloid leukemia at diagnosis by two-step RT-PCR. The sensitivity of the assay was 1:100 (first step) and 1:10 000 (second step), respectively. WT1-specific mRNA was detected in 73% of patients. No correlation was found between WT1 gene expression and age, FAB type, LDH and karyotype at diagnosis. All patients were treated with standard induction chemotherapy. There was no difference in the CR rate between WT1-positive and -negative patients. Using Kaplan and Meier plot analysis we found no difference in disease-free survival (DFS) and overall survival (OS) between patients displaying the WT1 transcript and WT1-negative patients. Furthermore, no significant interactions between WT1 PCR results and age, FAB type, LDH and karyotype on DFS and OS were demonstrable using Cox regression analysis. Eight patients who were WT1 PCR positive at diagnosis and achieved complete hematological remission following chemotherapy were monitored during the course of the disease. Based on our limited data demonstrating a heterogenity of WT1 PCR results in CR we cannot draw any conclusions regarding the usefulness of WT1 PCR analysis for the early detection of relapse. We conclude that WT1 gene expression at diagnosis is not associated with specific characteristics of AML blast cells and is not a prognostic factor for CR, remission duration and overall survival in acute myeloid leukemia.


Arthritis & Rheumatism | 1998

Expression of the C5a receptor (CD88) on synovial mast cells in patients with rheumatoid arthritis

Hans P. Kiener; Mehrdad Baghestanian; Martin Dominkus; Sabine Walchshofer; Minoo Ghannadan; Martin Willheim; Christian Sillaber; Winfried Graninger; Josef S Smolen; Peter Valent

OBJECTIVE To analyze the immunophenotype and functional properties of synovial mast cells (SyMC) in patients with rheumatoid arthritis (RA) and osteoarthritis (OA). METHODS Synovial tissue was obtained from 25 patients with RA and 17 patients with OA. Tissue was dispersed by enzymatic digestion using collagenase. Surface receptor expression on SyMC was analyzed by monoclonal antibodies (MAb) and indirect immunofluorescence staining. Histamine release experiments were performed using the MC agonist recombinant human (rHu) stem cell factor (SCF), the anaphylatoxin rHuC5a, and an anti-IgE antibody. RESULTS In both groups of patients (RA and OA), SyMC were found to react with MAb to IgE, SCF receptor (c-kit, CD117), as well as CD antigens likewise expressed in lung MC (CD9, CD29, CD33, CD43, CD44, CD45). However, a significantly increased proportion of SyMC from RA patients reacted with MAb against C5a receptor (C5aR; CD88), compared with SyMC from OA (mean +/- SD percentage of SyMC reacting with CD88 MAb S5/1 in RA 27.5 +/- 8.6% versus 0.0% in OA, and with CD88 MAb W17/1 in RA 58.3 +/- 15.2% versus 12.5 +/- 15.0% in OA; P < 0.05). Furthermore, in RA, significant histamine release from SyMC above control was induced by rHuC5a, anti-IgE, and rHuSCF, whereas SyMC in OA released histamine after stimulation with anti-IgE and rHuSCF, but not rHuC5a. CONCLUSION SyMC exhibit phenotypic and functional properties similar to MC in other tissues. In patients with RA, but not OA, SyMC express significant amounts of C5aR (CD88) and release histamine in response to rHuC5a. These results indicate a role for SyMC and C5a/C5aR in the pathogenesis of RA.


Haematologica | 2007

Synergistic growth-inhibitory effects of two tyrosine kinase inhibitors, dasatinib and PKC412, on neoplastic mast cells expressing the D816V-mutated oncogenic variant of KIT

Karoline V. Gleixner; Matthias Mayerhofer; Karoline Sonneck; Alexander Gruze; Puchit Samorapoompichit; Christian Baumgartner; Francis Y. Lee; Karl J. Aichberger; Paul W. Manley; Doriano Fabbro; Winfried F. Pickl; Christian Sillaber; Peter Valent

Background and Objectives In a majority of all patients with systemic mastocytosis (SM) including those with mast cell leukemia (MCL), neoplastic mast cells (MC) display the D816V-mutated variant of KIT. The respective oncoprotein, KIT D816V, exhibits constitutive tyrosine kinase (TK) activity and has been implicated in malignant cell growth. Therefore, several attempts have been made to identify KIT D816V-targeting drugs. Design and Methods We examined the effects of the novel TK-inhibitor dasatinib alone and in combination with other targeted drugs on growth of neoplastic MC. Results Confirming previous studies, dasatinib was found to inhibit the TK activity of wild type (wt) KIT and KIT-D816V as well as growth and survival of neoplastic MC and of the MCL cell line, HMC-1. The growth-inhibitory effects of dasatinib in HMC-1 cells were found to be associated with a decrease in expression of CD2 and CD63. In addition, we found that dasatinib blocks KIT D816V-induced cluster-formation and viability in Ba/F3 cells. In drug combination experiments, dasatinib was found to co-operate with PKC412, AMN107, imatinib, and 2CdA in producing growth-inhibition and apoptosis in neoplastic MC. In HMC-1.1 cells lacking KIT D816V, all drug interactions were found to be synergistic in nature. By contrast, in HMC-1.2 cells exhibiting KIT D816V, only the combinations dasatinib+PKC412 and dasatinib+2CdA were found to produce synergistic effects. Interpretation and Conclusions Combinations of targeted drugs may represent an interesting pharmacologic approach for the treatment of aggressive SM or MCL.


European Journal of Clinical Investigation | 2009

Immunosuppression and atypical infections in CML patients treated with dasatinib at 140 mg daily

Christian Sillaber; Harald Herrmann; Keiryn L. Bennett; Uwe Rix; Christian Baumgartner; Alexandra Böhm; Susanne Herndlhofer; Erwin Tschachler; Giulio Superti-Furga; Ulrich Jäger; Peter Valent

Background  The multikinase inhibitor dasatinib exerts growth‐inhibitory effects in patients with imatinib‐resistant chronic myeloid leukaemia (CML). In first clinical trials, side effects of dasatinib, 140 mg daily, were reported to be mild and tolerable.


Journal of Clinical Immunology | 2002

Cross-Priming of Cytotoxic T Cells Promoted by Apoptosis-Inducing Tumor Cell Reactive Antibodies?

Nicole Selenko; Otto Majdic; Ulrich Jäger; Christian Sillaber; Johannes Stöckl; Walter Knapp

Humanizing xenogenic monoclonal antibodies (MAbs) by genetic engineering has greatly improved their therapeutic utility and efficacy. The chimeric CD20 MAb C2B8 (Rituximab) is a prominent representative of this new generation of therapeutic MAbs and has been proposed as a treatment of choice for recurrent follicular non-Hodgkins lymphomas. Treatment of CD20+ B cells with MAb C2B8 triggers several cell-damaging actions including complement-mediated lysis (CDL), antibody-dependent cellular cytotoxicity (ADCC), and MAb-induced induction of apoptosis. We provide an overview of the most prominent mechanisms underlying the efficacy of antibody treatment. We introduce our concept of cross-priming of cytotoxic T-cell responses promoted by apoptosis incucing antibodies. Treatment of tumor cells with antibodies that are capable of inducing a proapoptotic signal via their cell surface target structure may not only contribute to their direct killing but also may induce cellular responses against the tumor, which may have a long-lasting protective effect. We report, using the example of C2B8 anti-CD20 treatment of lymphoma cells, that MAb C2B8-induced apoptosis of lymphoma cells not only kills these cells but also promotes uptake and cross-presentation of lymphoma cell-derived peptides by antigen-presenting dendritic cells (DC), induces maturation of DC, and allows the generation of specific CTL.


The FASEB Journal | 2005

Identification of mTOR as a novel bifunctional target in chronic myeloid leukemia: dissection of growth-inhibitory and VEGF-suppressive effects of rapamycin in leukemic cells

Matthias Mayerhofer; Karl J. Aichberger; Stefan Florian; Maria Theresa Krauth; Alexander W. Hauswirth; Sophia Derdak; Wolfgang R. Sperr; Harald Esterbauer; Oswald Wagner; Christine Marosi; Winfried F. Pickl; Michael W. Deininger; Ellen Weisberg; Brian J. Druker; James D. Griffin; Christian Sillaber; Peter Valent

The mammalian target of rapamycin (mTOR) has recently been described to be constitutively activated in Bcr‐Abl‐transformed cells and to mediate rapamycin‐induced inhibition of growth in respective cell lines. We have recently shown that rapamycin down‐regulates expression of vascular endothelial growth factor (VEGF), a mediator of leukemia‐associated angiogenesis, in primary CML cells. In the present study, we analyzed growth‐inhibitory in vitro and in vivo effects of rapamycin on primary CML cells and asked whether rapamycin‐induced suppression of VEGF in leukemic cells is related to growth inhibition. Rapamycin dose dependently inhibited growth of primary CML cells obtained from patients with imatinib‐responsive or imatinib‐resistant disease as well as growth of Bcr‐Abl‐transformed imatinib‐resistant cell lines. Moreover, we observed potent cytoreductive effects of rapamycin in a patient with imatinib‐resistant Bcr‐Abl+ leukemia. The growth‐inhibitory effects of rapamycin on CML cells were found to be associated with G1 cell cycle arrest and with induction of apoptosis. In all cell types tested, rapamycin was found to down‐regulate expression of VEGF. However, exogenously added VEGF did not counteract the rapamycin‐induced decrease in proliferation. In conclusion, rapamycin inhibits growth of CML cells in vitro and in vivo and, in addition, down‐regulates expression of VEGF. Both effects may contribute to the antileukemic activity of the drug in CML.


Journal of Immunology | 2008

Unique Effects of KIT D816V in BaF3 Cells: Induction of Cluster Formation, Histamine Synthesis, and Early Mast Cell Differentiation Antigens

Matthias Mayerhofer; Karoline V. Gleixner; Andrea Hoelbl; Stefan Florian; Gregor Hoermann; Karl J. Aichberger; Martin Bilban; Harald Esterbauer; Maria-Theresa Krauth; Wolfgang R. Sperr; Jack B. Longley; Robert Kralovics; Richard Moriggl; Jacques Zappulla; Roland S. Liblau; Ilse Schwarzinger; Veronika Sexl; Christian Sillaber; Peter Valent

Oncogenic tyrosine kinases (TK) usually convert growth factor-dependent cells to factor independence with autonomous proliferation. However, TK-driven neoplasms often are indolent and characterized by cell differentiation rather than proliferation. A prototype of an indolent TK-driven neoplasm is indolent systemic mastocytosis. We found that the D816V-mutated variant of KIT, a TK detectable in most patients with systemic mastocytosis, induces cluster formation and expression of several mast cell differentiation and adhesion Ags, including microphthalmia transcription factor, IL-4 receptor, histamine, CD63, and ICAM-1 in IL-3-dependent BaF3 cells. By contrast, wild-type KIT did not induce cluster formation or mast cell differentiation Ags. Additionally, KIT D816V, but not wild-type KIT, induced STAT5 activation in BaF3 cells. However, despite these intriguing effects, KIT D816V did not convert BaF3 cells to factor-independent proliferation. Correspondingly, BaF3 cells with conditional expression of KIT D816V did not form tumors in nude mice. Together, the biologic effects of KIT D816V in BaF3 cells match strikingly with the clinical course of indolent systemic mastocytosis and with our recently established transgenic mouse model, in which KIT D816V induces indolent mast cell accumulations but usually does not induce a malignant mast cell disease. Based on all these results, it is hypothesized that KIT D816V as a single hit may be sufficient to cause indolent systemic mastocytosis, whereas additional defects may be required to induce aggressive mast cell disorders.

Collaboration


Dive into the Christian Sillaber's collaboration.

Top Co-Authors

Avatar

Peter Valent

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Matthias Mayerhofer

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Klaus Lechner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Winfried F. Pickl

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Karl J. Aichberger

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

Karoline V. Gleixner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Alexander Gruze

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Maria-Theresa Krauth

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Harald Esterbauer

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge