Alexander Egle
Seattle Children's Research Institute
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Alexander Egle.
Cancer Cell | 2013
Gloria Lutzny; Thomas Kocher; Marc Schmidt-Supprian; Martina Rudelius; Ludger Klein-Hitpass; Andrew J. Finch; Jan Dürig; Michaela Wagner; Claudia Haferlach; Alexander Kohlmann; Susanne Schnittger; Marc Seifert; Stefan Wanninger; Nadja Zaborsky; Robert A.J. Oostendorp; Jürgen Ruland; Michael Leitges; Toni Kuhnt; Yvonne Schäfer; Benedikt Lampl; Christian Peschel; Alexander Egle; Ingo Ringshausen
Summary Tumor cell survival critically depends on heterotypic communication with benign cells in the microenvironment. Here, we describe a survival signaling pathway activated in stromal cells by contact to B cells from patients with chronic lymphocytic leukemia (CLL). The expression of protein kinase C (PKC)-βII and the subsequent activation of NF-κB in bone marrow stromal cells are prerequisites to support the survival of malignant B cells. PKC-β knockout mice are insusceptible to CLL transplantations, underscoring the in vivo significance of the PKC-βII-NF-κB signaling pathway in the tumor microenvironment. Upregulated stromal PKC-βII in biopsies from patients with CLL, acute lymphoblastic leukemia, and mantle cell lymphoma suggests that this pathway may commonly be activated in a variety of hematological malignancies.
Leukemia | 2016
Andy C. Rawstron; C. Fazi; Andreas Agathangelidis; Neus Villamor; R. Letestu; Josep Nomdedeu; C. Palacio; Olga Stehlíková; Karl-Anton Kreuzer; S. Liptrot; D. OBrien; R de Tute; I. Marinov; M. Hauwel; Martin Spacek; J. Dobber; Arnon P. Kater; Peter Gambell; Asha Soosapilla; Gerard Lozanski; G. Brachtl; Ke Lin; Justin Boysen; Curtis A. Hanson; Jeffrey L. Jorgensen; Maryalice Stetler-Stevenson; Constance Yuan; H. E. Broome; Laura Z. Rassenti; F. Craig
In chronic lymphocytic leukemia (CLL) the level of minimal residual disease (MRD) after therapy is an independent predictor of outcome. Given the increasing number of new agents being explored for CLL therapy, using MRD as a surrogate could greatly reduce the time necessary to assess their efficacy. In this European Research Initiative on CLL (ERIC) project we have identified and validated a flow-cytometric approach to reliably quantitate CLL cells to the level of 0.0010% (10−5). The assay comprises a core panel of six markers (i.e. CD19, CD20, CD5, CD43, CD79b and CD81) with a component specification independent of instrument and reagents, which can be locally re-validated using normal peripheral blood. This method is directly comparable to previous ERIC-designed assays and also provides a backbone for investigation of new markers. A parallel analysis of high-throughput sequencing using the ClonoSEQ assay showed good concordance with flow cytometry results at the 0.010% (10−4) level, the MRD threshold defined in the 2008 International Workshop on CLL guidelines, but it also provides good linearity to a detection limit of 1 in a million (10−6). The combination of both technologies would permit a highly sensitive approach to MRD detection while providing a reproducible and broadly accessible method to quantify residual disease and optimize treatment in CLL.
Leukemia | 2016
Bola Hanna; Fabienne McClanahan; H Yazdanparast; Nadja Zaborsky; Verena Kalter; P M Rößner; Axel Benner; Claudia Dürr; Alexander Egle; John G. Gribben; Peter Lichter; Martina Seiffert
Chronic lymphocytic leukemia (CLL) is characterized by apoptosis resistance and a dysfunctional immune system. Previous reports suggested a potential role of myeloid cells in mediating these defects. However, the composition and function of CLL-associated myeloid cells have not been thoroughly investigated in vivo. Using the Eμ-TCL1 mouse model, we observed severe skewing of myeloid cell populations with CLL development. Monocytes and M2-like macrophages infiltrated the peritoneal cavity of leukemic mice. Monocytes also accumulated in the spleen in a CCR2-dependent manner, and were severely skewed toward Ly6Clow patrolling or nonclassical phenotype. In addition, the percentage of MHC-IIhi dendritic cells and macrophages significantly dropped in the spleen. Gene expression profiling of CLL-associated monocytes revealed aberrantly high PD-L1 expression and secretion of multiple inflammatory and immunosuppressive cytokines like interleukin-10, tumor necrosis factor-α and CXCL9. In vivo myeloid cell depletion using liposomal Clodronate resulted in a significant control of CLL development accompanied by a pronounced repair of innate immune cell phenotypes and a partial resolution of systemic inflammation. In addition, CLL-associated skewing of T cells toward antigen-experienced phenotypes was repaired. The presented data suggest that targeting nonmalignant myeloid cells might serve as a novel immunotherapeutical strategy for CLL.
The Lancet Haematology | 2016
Richard Greil; Petra Obrtlikova; Lukas Smolej; Tomas Kozak; Michael Steurer; Johannes Andel; Sonja Burgstaller; Eva Mikuskova; Liana Gercheva; Thomas Nösslinger; Tomáš Papajík; Miriam Ladická; Michael Girschikofsky; Mikuláš Hrubiško; Ulrich Jäger; Michael A. Fridrik; Martin Pecherstorfer; Eva Králiková; Cristina Burcoveanu; Emil Spasov; Andreas L. Petzer; Georgi Mihaylov; Julian Raynov; Horst Oexle; August Zabernigg; Emília Flochová; Stanislav Palášthy; Olga Stehlíková; Michael Doubek; Petra Altenhofer
BACKGROUND In many patients with chronic lymphocytic leukaemia requiring treatment, induction therapy with rituximab plus chemotherapy improves outcomes compared with chemotherapy alone. In this study we aimed to investigate the potential of rituximab maintenance therapy to prolong disease control in patients who respond to rituximab-containing induction regimens. METHODS In this randomised, international, multicentre, open-label, phase 3 clinical trial, we enrolled patients who had achieved a complete response (CR), CR with incomplete bone marrow recovery (CRi), or partial response (PR) to first-line or second-line rituximab-containing chemoimmunotherapy and randomly assigned them in a 1:1 ratio (central block randomisation in the electronic case report form system) to either intravenous rituximab 375 mg/m(2) every 3 months, or observation alone, for 2 years. Stratification was by country, line of treatment, type of chemotherapy added to the rituximab backbone, and degree of remission following induction. The primary endpoint was progression-free survival. Efficacy analysis was done in the intention-to-treat population. This is the final, event-triggered analysis. Final analysis was triggered by the occurrence of 92 events. This trial is registered with ClinicalTrials.gov, number NCT01118234. FINDINGS Between April 1, 2010, and Dec 23, 2013, 134 patients were randomised to rituximab and 129 to observation alone. Median observation times were 33·4 months (IQR 25·7-42·8) for the rituximab group and 34·0 months (25·4-41·9) for the observation group. Progression-free survival was significantly longer in the rituximab maintenance group (47·0 months, IQR 28·5-incalculable) than with observation alone (35·5 months, 95% CI 25·7-46·3; hazard ratio [HR] 0·50, 95% CI 0·33-0·75, p=0·00077). The incidence of grade 3-4 haematological toxicities other than neutropenia was similar in the two treatment groups. Grade 3-4 neutropenia occurred in 28 (21%) patients in the rituximab group and 14 (11%) patients in the observation group. Apart from neutropenia, the most common grade 3-4 adverse events were upper (five vs one [1%] patient in the observation group) and lower (three [2%] vs one [1%]) respiratory tract infection, pneumonia (nine [7%] vs two [2%]), thrombopenia (four [3%] vs four [3%]), neoplasms (five [4%] vs four [3%]), and eye disorders (four [3%] vs two [2%]). The overall incidence of infections of all grades was higher among rituximab recipients (88 [66%] vs 65 [50%]). INTERPRETATION Rituximab maintenance therapy prolongs progression-free survival in patients achieving at least a PR to induction with rituximab plus chemotherapy, and the treatment is well tolerated overall. Although it is associated with an increase in infections, there is no excess in infection mortality, suggesting that remission maintenance with rituximab is an effective and safe option in the management of chronic lymphocytic leukaemia in early treatment phases. FUNDING Arbeitsgemeinschaft Medikamentöse Tumortherapie gemeinnützige GmbH (AGMT), Roche.
OncoImmunology | 2018
Kemal Catakovic; Franz Josef Gassner; Christoph Ratswohl; Nadja Zaborsky; Stefan Rebhandl; Maria Schubert; Markus Steiner; Julia Christine Gutjahr; Lisa Pleyer; Alexander Egle; Tanja Nicole Hartmann; Richard Greil; Roland Geisberger
ABSTRACT While research on T cell exhaustion in context of cancer particularly focuses on CD8+ cytotoxic T cells, the role of inhibitory receptors on CD4+ T-helper cells have remained largely unexplored. TIGIT is a recently identified inhibitory receptor on T cells and natural killer (NK) cells. In this study, we examined TIGIT expression on T cell subsets from CLL patients. While we did not observe any differences in TIGIT expression in CD8+ T cells of healthy controls and CLL cells, we found an enrichment of TIGIT+ T cells in the CD4+ T cell compartment in CLL. Intriguingly, CLL patients with an advanced disease stage displayed elevated numbers of CD4+ TIGIT+ T cells compared to low risk patients. Autologous CLL-T cell co-culture assays revealed that depleting CD4+ TIGIT+ expressing T cells from co-cultures significantly decreased CLL viability. Accordingly, a supportive effect of TIGIT+CD4+ T cells on CLL cells in vitro could be recapitulated by blocking the interaction of TIGIT with its ligands using TIGIT-Fc molecules, which also impeded the T cell specific production of CLL-prosurvival cytokines. Our data reveal that TIGIT+CD4+T cells provide a supportive microenvironment for CLL cells, representing a potential therapeutic target for CLL treatment.
Leukemia | 2018
Nadja Zaborsky; Franz Josef Gassner; Jan P. Höpner; Maria Schubert; Daniel Hebenstreit; Richard Stark; Daniela Asslaber; Markus Steiner; Roland Geisberger; Richard Greil; Alexander Egle
The TCL1 mouse model is widely used to study pathophysiology, clonal evolution, and drug sensitivity or resistance of chronic lymphocytic leukemia (CLL). By performing whole exome sequencing, we present the genetic landscape of primary tumors from TCL1 mice and of TCL1 tumors serially transplanted into wild-type recipients to mimic clonal evolution. We show that similar to CLL patients, mutations in mice are frequently subclonal and heterogenous among different primary TCL1 mice. We further describe that this molecular heterogeneity mirrors heterogenous disease characteristics such as organ infiltration or CLL dependent T cell skewing. Similar to human CLL, we further observed the occurrence of novel mutations and structural variations during clonal evolution and found plasticity in the expansion of B cell receptor specific subclones. Thus, our results uncover that the genetic complexity, pathway dependence and clonal dynamics in mouse CLL are in relevant agreement to human CLL, and they are important to consider in future research using the TCL1 mouse for studying CLL.
BMC Cancer | 2015
Thomas Melchardt; Clemens Hufnagl; Teresa Magnes; Lukas Weiss; Georg Hutarew; Daniel Neureiter; Alexander Schlattau; Gerhard Moser; Alexander Gaggl; Wolfgang Tränkenschuh; Richard Greil; Alexander Egle
Blood | 2014
Bola Hanna; Fabienne McClanahan; Nadja Zaborsky; Claudia Dürr; Verena Kalter; Alexander Egle; John G. Gribben; Peter Lichter; Martina Seiffert
Future Oncology | 2018
Michael Leisch; Alexander Egle; Richard Greil
Annals of Hematology | 2018
Alexander Egle; Michael Steurer; Thomas Melchardt; Lukas Weiss; Franz Josef Gassner; Nadja Zaborsky; Roland Geisberger; Kemal Catakovic; Tanja Nicole Hartmann; Lisa Pleyer; Daniela Voskova; Josef Thaler; Alois Lang; Michael Girschikofsky; Andreas L. Petzer; Richard Greil