Network


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

Hotspot


Dive into the research topics where Eugen Leo is active.

Publication


Featured researches published by Eugen Leo.


Science | 2008

Tumor Regression in Cancer Patients by Very Low Doses of a T Cell–Engaging Antibody

Ralf Bargou; Eugen Leo; Gerhard Zugmaier; Matthias Klinger; Mariele Goebeler; Stefan Knop; Richard Noppeney; Andreas Viardot; Georg Hess; Martin Schuler; Hermann Einsele; Christian Brandl; A. Wolf; Petra Kirchinger; Petra Klappers; Margit Schmidt; Gert Riethmüller; Carsten Reinhardt; Patrick A. Baeuerle; Peter Kufer

Previous attempts have shown the potential of T cells in immunotherapy of cancer. Here, we report on the clinical activity of a bispecific antibody construct called blinatumomab, which has the potential to engage all cytotoxic T cells in patients for lysis of cancer cells. Doses as low as 0.005 milligrams per square meter per day in non–Hodgkins lymphoma patients led to an elimination of target cells in blood. Partial and complete tumor regressions were first observed at a dose level of 0.015 milligrams, and all seven patients treated at a dose level of 0.06 milligrams experienced a tumor regression. Blinatumomab also led to clearance of tumor cells from bone marrow and liver. T cell–engaging antibodies appear to have therapeutic potential for the treatment of malignant diseases.


Journal of Clinical Oncology | 2016

Bispecific T-Cell Engager (BiTE) Antibody Construct Blinatumomab for the Treatment of Patients With Relapsed/Refractory Non-Hodgkin Lymphoma: Final Results From a Phase I Study

Maria-Elisabeth Goebeler; Stefan Knop; Andreas Viardot; Peter Kufer; Max S. Topp; Hermann Einsele; Richard Noppeney; Georg Hess; Stefan Kallert; Andreas Mackensen; Kathrin Rupertus; Lothar Kanz; Martin Libicher; Dirk Nagorsen; Gerhard Zugmaier; Matthias Klinger; A. Wolf; Brigitte Dorsch; Beate D. Quednau; Margit Schmidt; Jürgen Scheele; Patrick A. Baeuerle; Eugen Leo; Ralf C. Bargou

PURPOSE Blinatumomab is a CD19/CD3 BiTE (bispecific T-cell engager) antibody construct for the treatment of Philadelphia chromosome-negative acute B-lymphoblastic leukemia. We evaluated blinatumomab in relapsed/refractory B-cell non-Hodgkin lymphoma (NHL). PATIENTS AND METHODS This 3 + 3 design, phase I dose-escalation study determined adverse events and the maximum tolerated dose (MTD) of continuous intravenous infusion blinatumomab in patients with relapsed/refractory NHL. Blinatumomab was administered over 4 or 8 weeks at seven different dose levels (0.5 to 90 μg/m(2)/day). End points were incidence of adverse events, pharmacokinetics, pharmacodynamics, and overall response rate. RESULTS Between 2004 and 2011, 76 heavily pretreated patients with relapsed/refractory NHL, who included 14 with diffuse large B-cell lymphoma, were enrolled; 42 received treatment in the formal dose-escalation phase. Neurologic events were dose limiting, and 60 μg/m(2)/day was established as the MTD. Thirty-four additional patients were recruited to evaluate antilymphoma activity and strategies for mitigating neurologic events at a prespecified MTD. Stepwise dosing (5 to 60 μg/m(2)/day) plus pentosan polysulfate SP54 (n = 3) resulted in no treatment discontinuations; single-step (n = 5) and double-step (n = 24) dosing entailed two and seven treatment discontinuations due to neurologic events, respectively. Grade 3 neurologic events occurred in 22% of patients (no grade 4/5). Among patients treated at 60 μg/m(2)/day (target dose; n = 35), the overall response rate was 69% across NHL subtypes and 55% for diffuse large B-cell lymphoma (n = 11); median response duration was 404 days (95% CI, 207 to 1,129 days). CONCLUSION In this phase I study of relapsed/refractory NHL, continuous infusion with CD19-targeted immunotherapy blinatumomab at various doses and schedules was feasible, with an MTD of 60 μg/m(2)/day. Single-agent blinatumomab showed antilymphoma activity.


British Journal of Haematology | 2010

Preclinical anti-myeloma activity of the novel HDAC-inhibitor JNJ-26481585

Thorsten Stühmer; Janine Arts; Manik Chatterjee; Johanna Borawski; André Wolff; Peter H. King; Hermann Einsele; Eugen Leo; Ralf C. Bargou

Pharmacological inhibitors of histone deacetylases (HDACs) are currently being developed and tested as anti‐cancer agents and may be useful to enhance the therapeutic efficiency of established anti‐myeloma treatments. This study preclinically evaluated the effects of the ‘second generation’ pan‐HDAC inhibitor JNJ‐26481585 on human multiple myeloma (MM) cells from established cell lines and primary MM samples (n = 42). Molecular responses in both groups of MM cells included histone acetylation, a shift in Bcl2‐family members towards proapoptotic bias, attenuation of growth and survival pathway activity and Hsp72 induction. Mcl‐1 depletion and Hsp72 induction were the most reliable features observed in JNJ‐26481585‐treated primary MM samples. The drug alone effectively induced myeloma cell death at low nanomolar concentrations. In vitro combination of JNJ‐26481585 with anti‐myeloma therapeutic agents generally resulted In effects close to additivity. In view of the favourable activity of this novel HDAC‐inhibitor towards primary myeloma cells further evaluation in a clinical setting is warranted.


Oncotarget | 2015

TGF-ß isoforms in cancer: Immunohistochemical expression and Smad-pathway-activity-analysis in thirteen major tumor types with a critical appraisal of antibody specificity and immunohistochemistry assay validity.

Markus J. Riemenschneider; Maria Hirblinger; Arabel Vollmann-Zwerenz; Peter Hau; Martin Proescholdt; Frank Jaschinski; Tanja Rothhammer-Hampl; Katja Wosikowski; Michel Janicot; Eugen Leo

The literature on TGF-Δ in cancer including data on the expression or activation of TGF-Δ pathway components in specific tumors types is steadily growing. However, no systematic and uniform analysis exists reporting expression levels of the main TGF-Δ pathway components across the most frequent tumor types. We used a standardized immunohistochemical assay investigating TGF-Δ isoform expression and pathway activation across 13 different tumor types and corresponding non-neoplastic tissues. The study was performed on tissue microarrays allowing for the parallel analysis of a total of 1638 human tumor samples. TGF-Δ1, TGF-Δ2 and p-Smad2/3 were substantially expressed in multiple cancers widening the options for TGF-Δ isoform directed therapies. Of note, TGF-Δ antigens appear to be expressed in an individual manner pointing towards a need for patient preselection for TGF-β isoform specific treatment. Yet, a thorough investigation of antibody specificity and assay validity revealed that immunohistochemistry did not correlate with other detection methods on mRNA or protein level in all instances. As such, with the currently available means (i.e. antibodies tested) a stratification of patients within clinical trials for TGF-Δ directed antisense therapies based upon TGF-β immunohistochemistry alone has to be interpreted with caution and should be carefully evaluated in combination with other parameters.


PLOS ONE | 2017

First-in-human phase I study of ISTH0036, an antisense oligonucleotide selectively targeting transforming growth factor beta 2 (TGF-β2), in subjects with open-angle glaucoma undergoing glaucoma filtration surgery

Norbert Pfeiffer; Bogomil Voykov; Giulia Renieri; Katharina Bell; Paul Richter; Melanie Weigel; Hagen Thieme; Barbara Wilhelm; Katrin Lorenz; Martin Feindor; Katja Wosikowski; Michel Janicot; Daniela Päckert; Regina Römmich; Carola Mala; Petra Fettes; Eugen Leo

Purpose To evaluate the safety and tolerability of intravitreal ISTH0036, an antisense oligonucleotide selectively targeting transforming growth factor beta 2 (TGF-β2), in patients with primary open angle glaucoma (POAG) undergoing trabeculectomy (TE; glaucoma filtration surgery). Methods In this prospective phase I trial glaucoma patients scheduled for TE with mitomycin C (MMC) received a single intravitreal injection of ISTH0036 at the end of surgery in escalating total doses of 6.75 μg, 22.5 μg, 67.5 μg or 225 μg, resulting in calculated intraocular ISTH0036 concentrations in the vitreous humor of approximately 0.3 μM, 1 μM, 3 μM or 10 μM after injection, respectively. Outcomes assessed included: type and frequency of adverse events (AEs), intraocular pressure (IOP), numbers of interventions post trabeculectomy, bleb survival, visual acuity, visual field, electroretinogram (ERG), slit lamp biomicroscopy and optic disc assessment. Results In total, 12 patients were treated in the 4 dose groups. Main ocular AEs observed were corneal erosion, corneal epithelium defect, or too high or too low IOP, among others. No AE was reported to be related to ISTH0036. All other safety-related analyses did not reveal any toxicities of concern, either. The mean medicated preoperative IOP at decision time-point for surgery was 27.3 mmHg +/- 12.6 mmHg (SD). Mean IOP (±SD) for dose levels 1, 2, 3, and 4 were at Day 43 9.8 mmHg ± 1.0 mmHg, 11.3 mmHg ± 6.7 mmHg, 5.5 mmHg ± 3.0 mmHg and 7.5 mmHg ± 2.3 mmHg SD; and at Day 85 9.7 mmHg ± 3.3 mmHg, 14.2 mmHg ± 6.5 mmHg, 5.8 mmHg ± 1.8 mmHg and 7.8 mmHg ± 0.6 mmHg, respectively. In contrast to IOP values for dose levels 1 and 2, IOP values for dose levels 3 and 4 persistently remained below 10 mmHg throughout the observation period. Conclusion This first-in-human trial demonstrates that intravitreal injection of ISTH0036 at the end of TE is safe. Regarding IOP control, single-dose ISTH0036 administration of 67.5 μg or 225 μg at the time of TE resulted in IOP values persistently < 10 mmHg over the three month postoperative observation period.


Cancer Immunology, Immunotherapy | 2006

T-cell activation and B-cell depletion in chimpanzees treated with a bispecific anti-CD19/anti-CD3 single-chain antibody construct

Bernd Schlereth; Cornelia Quadt; Torsten Dreier; Peter Kufer; Grit Lorenczewski; Nadja Prang; Christian Brandl; Sandra Lippold; Kathy E. Cobb; Kathleen M. Brasky; Eugen Leo; Ralf C. Bargou; Krishna K. Murthy; Patrick A. Baeuerle


European Journal of Cancer | 2006

A phase I study with adecatumumab, a human antibody directed against epithelial cell adhesion molecule, in hormone refractory prostate cancer patients

Ralf Oberneder; Dorothea Weckermann; Beatrice Ebner; Cornelia Quadt; Petra Kirchinger; Tobias Raum; Mathias Locher; Nadja Prang; Patrick A. Baeuerle; Eugen Leo


Hepatology Research | 2006

Expression of epithelial cellular adhesion molecule (Ep-CAM) in chronic (necro-)inflammatory liver diseases and hepatocellular carcinoma

Kai Breuhahn; Patrick A. Baeuerle; Malte Peters; Nadja Prang; U. Töx; Rudolph Köhne-Volland; Volker Dries; Peter Schirmacher; Eugen Leo


Cancer Research | 2008

JNJ-26854165 - a novel hdm2 antagonist in clinical development showing broad-spectrum preclinical antitumor activity against solid malignancies

Janine Arts; Martin John Page; Annemie Francine Valckx; Christine Blattner; R. Kulikov; Wim Floren; Luc Van Nuffel; Lut Janssen; Peter H. King; Stefan Masure; Karine Smans; Peter Verhasselt; Hinrich Goehlmann; Yannick Aimé Eddy Ligny; Delphine Yvonne Raymonde Lardeau; Imre Csoka; Luc Andries; Dirk Brehmer; Eddy Jean Edgard Freyne; Eugen Leo; Michel Janicot; Christophe Meyer; Jean Fernand Armand Lacrampe; Bruno Schoentjes


Journal of Clinical Oncology | 2008

A first-in-class HDM2-inhibitor (JNJ-26854165) in phase I development shows potent activity against multiple myeloma (MM) cells in vitro and ex vivo

T. Stühmer; J. Arts; P. King; M. Page; K. Bommert; Eugen Leo; Ralf C. Bargou

Collaboration


Dive into the Eugen Leo's collaboration.

Top Co-Authors

Avatar

Ralf C. Bargou

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Katja Wosikowski

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge