Network


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

Hotspot


Dive into the research topics where Matthias Mayerhofer is active.

Publication


Featured researches published by Matthias Mayerhofer.


The American Journal of Surgical Pathology | 2004

CD25 indicates the neoplastic phenotype of mast cells: a novel immunohistochemical marker for the diagnosis of systemic mastocytosis (SM) in routinely processed bone marrow biopsy specimens.

Karl Sotlar; Hans-Peter Horny; Ingrid Simonitsch; Manuela Krokowski; Karl J. Aichberger; Matthias Mayerhofer; Dieter Printz; Gerhard Fritsch; Peter Valent

The diagnosis of systemic mastocytosis (SM) is based primarily on the histologic and immunohistochemical evaluation of a bone marrow trephine biopsy specimen. Although mast cell (MC) specific antigens like tryptase and chymase are detectable in routinely processed tissue, no immunohistochemical markers that can be used to discriminate between normal and neoplastic MCs are yet available. We have investigated the diagnostic value of an antibody against CD25 for the immunohistochemical detection of MCs in bone marrow sections in 73 patients with SM and 75 control cases (reactive marrow, n = 54; myelogenous neoplasms, n = 21) and correlated the results with the presence of c-kit mutations. While MCs in almost all patients with SM (72 of 73) expressed CD25, none of the control samples contained CD25-positive MCs. Irrespective of the SM subtype, most of neoplastic MCs expressed CD25. In 3 patients with advanced MC disease, pure populations of neoplastic MCs were obtained and found to express CD25 mRNA by RT-PCR analysis. In addition, all patients with CD25-positive MCs contained c-kit mutations, while all control cases exhibited wild type c-kit. CD25 therefore appears to be a reliable immunohistochemical marker for the discrimination of neoplastic from normal/reactive MCs, with potential as a diagnostic tool in SM.


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.


Cancer Cell | 2013

A Kinase-Independent Function of CDK6 Links the Cell Cycle to Tumor Angiogenesis.

Karoline Kollmann; Gerwin Heller; Christine Schneckenleithner; Wolfgang Warsch; Ruth Scheicher; Rene G. Ott; Markus Schäfer; Sabine Fajmann; Michaela Schlederer; Ana-Iris Schiefer; Ursula Reichart; Matthias Mayerhofer; Christoph Hoeller; Sabine Zöchbauer-Müller; Dontscho Kerjaschki; Christoph Bock; Lukas Kenner; Gerald Hoefler; Michael Freissmuth; Anthony R. Green; Richard Moriggl; Meinrad Busslinger; Marcos Malumbres; Veronika Sexl

Summary In contrast to its close homolog CDK4, the cell cycle kinase CDK6 is expressed at high levels in lymphoid malignancies. In a model for p185BCR-ABL+ B-acute lymphoid leukemia, we show that CDK6 is part of a transcription complex that induces the expression of the tumor suppressor p16INK4a and the pro-angiogenic factor VEGF-A. This function is independent of CDK6’s kinase activity. High CDK6 expression thus suppresses proliferation by upregulating p16INK4a, providing an internal safeguard. However, in the absence of p16INK4a, CDK6 can exert its full tumor-promoting function by enhancing proliferation and stimulating angiogenesis. The finding that CDK6 connects cell-cycle progression to angiogenesis confirms CDK6’s central role in hematopoietic malignancies and could underlie the selection pressure to upregulate CDK6 and silence p16INK4a.


Expert Review of Hematology | 2012

Pathogenesis and classification of eosinophil disorders: a review of recent developments in the field

Peter Valent; Gerald J. Gleich; Andreas Reiter; Florence Roufosse; Peter F. Weller; Andrzej Hellmann; Georgia Metzgeroth; Kristin M. Leiferman; Michel Arock; Karl Sotlar; Joseph H. Butterfield; Sabine Cerny-Reiterer; Matthias Mayerhofer; Peter Vandenberghe; Torsten Haferlach; Bruce S. Bochner; Jason Gotlib; Hans-Peter Horny; Hans-Uwe Simon; Amy D. Klion

Eosinophils and their products play an essential role in the pathogenesis of various reactive and neoplastic disorders. Depending on the underlying disease, molecular defect and involved cytokines, hypereosinophilia may develop and may lead to organ damage. In other patients, persistent eosinophilia is accompanied by typical clinical findings, but the causative role and impact of eosinophilia remain uncertain. For patients with eosinophil-mediated organ pathology, early therapeutic intervention with agents reducing eosinophil counts can be effective in limiting or preventing irreversible organ damage. Therefore, it is important to approach eosinophil disorders and related syndromes early by using established criteria, to perform all appropriate staging investigations, and to search for molecular targets of therapy. In this article, we review current concepts in the pathogenesis and evolution of eosinophilia and eosinophil-related organ damage in neoplastic and non-neoplastic conditions. In addition, we discuss classifications of eosinophil disorders and related syndromes as well as diagnostic algorithms and standard treatment for various eosinophil-related disorders.


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.


Blood | 2014

Dipeptidylpeptidase IV (CD26) defines leukemic stem cells (LSC) in chronic myeloid leukemia

Harald Herrmann; Irina Sadovnik; Sabine Cerny-Reiterer; Thomas Rülicke; Gabriele Stefanzl; Michael Willmann; Gregor Hoermann; Martin Bilban; Katharina Blatt; Susanne Herndlhofer; Matthias Mayerhofer; Berthold Streubel; Wolfgang R. Sperr; Tessa L. Holyoake; Christine Mannhalter; Peter Valent

Chronic myeloid leukemia (CML) is a stem cell (SC) neoplasm characterized by the BCR/ABL1 oncogene. Although mechanisms of BCR/ABL1-induced transformation are well-defined, little is known about effector-molecules contributing to malignant expansion and the extramedullary spread of leukemic SC (LSC) in CML. We have identified the cytokine-targeting surface enzyme dipeptidylpeptidase-IV (DPPIV/CD26) as a novel, specific and pathogenetically relevant biomarker of CD34(+)/CD38(─) CML LSC. In functional assays, CD26 was identified as target enzyme disrupting the SDF-1-CXCR4-axis by cleaving SDF-1, a chemotaxin recruiting CXCR4(+) SC. CD26 was not detected on normal SC or LSC in other hematopoietic malignancies. Correspondingly, CD26(+) LSC decreased to low or undetectable levels during successful treatment with imatinib. CD26(+) CML LSC engrafted NOD-SCID-IL-2Rγ(-/-) (NSG) mice with BCR/ABL1(+) cells, whereas CD26(─) SC from the same patients produced multilineage BCR/ABL1(-) engraftment. Finally, targeting of CD26 by gliptins suppressed the expansion of BCR/ABL1(+) cells. Together, CD26 is a new biomarker and target of CML LSC. CD26 expression may explain the abnormal extramedullary spread of CML LSC, and inhibition of CD26 may revert abnormal LSC function and support curative treatment approaches in this malignancy.


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.


Cancer Research | 2010

Polo-like Kinase 1 (Plk1) as a Novel Drug Target in Chronic Myeloid Leukemia: Overriding Imatinib Resistance with the Plk1 Inhibitor BI 2536

Karoline V. Gleixner; Veronika Ferenc; Barbara Peter; Alexander Gruze; Renata A. Meyer; Emir Hadzijusufovic; Sabine Cerny-Reiterer; Matthias Mayerhofer; Winfried F. Pickl; Christian Sillaber; Peter Valent

In most patients with chronic myeloid leukemia (CML), the disease can be kept under control using the BCR/ABL kinase inhibitor imatinib. Nevertheless, resistance or intolerance to imatinib and other BCR/ABL inhibitors may occur during therapy. Therefore, CML research is focusing on novel targets and targeted drugs. Polo-like kinase 1 (Plk1) is a serine/threonine kinase that plays an essential role in mitosis. In this study, we examined the expression of Plk1 in CML cells and its potential role as a therapeutic target. Plk1 was found to be expressed in phosphorylated form in the CML cell line K562 as well as in primary CML cells in all patients tested. Inhibition of BCR/ABL by imatinib or nilotinib (AMN107) led to decreased expression of the Plk1 protein in CML cells, suggesting that BCR/ABL promotes Plk1 generation. Silencing of Plk1 in CML cells by a small interfering RNA approach was followed by cell cycle arrest and apoptosis. Furthermore, the Plk1-targeting drug BI 2536 was found to inhibit proliferation of imatinib-sensitive and imatinib-resistant CML cells, including leukemic cells, carrying the T315 mutation of BCR/ABL with reasonable IC(50) values (1-50 nmol/L). The growth-inhibitory effects of BI 2536 on CML cells were found to be associated with cell cycle arrest and apoptosis. Moreover, BI 2536 was found to synergize with imatinib and nilotinib in producing growth inhibition in CML cells. In conclusion, Plk1 is expressed in CML cells and may represent a novel, interesting target in imatinib-sensitive and imatinib-resistant CML.

Collaboration


Dive into the Matthias Mayerhofer's collaboration.

Top Co-Authors

Avatar

Peter Valent

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Christian Sillaber

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

Winfried F. Pickl

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

Wolfgang R. Sperr

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Gregor Hoermann

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
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
Researchain Logo
Decentralizing Knowledge