Network


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

Hotspot


Dive into the research topics where Maxim Kebenko is active.

Publication


Featured researches published by Maxim Kebenko.


British Journal of Haematology | 2015

A phase I/II study of sunitinib and intensive chemotherapy in patients over 60 years of age with acute myeloid leukaemia and activating FLT3 mutations.

Walter Fiedler; Sabine Kayser; Maxim Kebenko; Melanie Janning; Jürgen Krauter; Marcus M. Schittenhelm; Katharina Götze; Daniela Weber; Gudrun Göhring; Veronica Teleanu; Felicitas Thol; Michael Heuser; Konstanze Döhner; Arnold Ganser; Hartmut Döhner; Richard F. Schlenk

Acute myeloid leukaemia (AML) with FLT3 mutation has a dismal prognosis in elderly patients. Treatment with a combination of FLT3 inhibitors and standard chemotherapy has not been extensively studied. Therefore, we instigated a phase I/II clinical trial of chemotherapy with cytosine arabinoside (Ara‐C)/daunorubicin induction (7+3) followed by three cycles of intermediate‐dose Ara‐C consolidation in 22 AML patients with activating FLT3 mutations. Sunitinib was added at predefined dose levels and as maintenance therapy for 2 years. At dose level 1, sunitinib 25 mg daily continuously from day 1 onwards resulted in two cases with dose‐limiting toxicity (DLT), prolonged haemotoxicity and hand‐foot syndrome. At dose level −1, sunitinib 25 mg was restricted to days 1–7 of each chemotherapy cycle. One DLT was observed in six evaluable patients. Six additional patients were treated in an extension phase. Thirteen of 22 patients (59%; 8/14 with FLT3–internal tandem duplication and 5/8 with FLT3‐tyrosine kinase domain) achieved a complete remission/complete remission with incomplete blood count recovery. For the 17 patients included at the lower dose level, median overall, relapse‐free and event‐free survival were 1·6, 1·0 and 0·4 years, respectively. Four out of five analysed patients with relapse during maintenance therapy lost their initial FLT3 mutation, suggesting outgrowth of FLT3 wild‐type subclones.


Cellular Signalling | 2015

ErbB2 signaling activates the Hedgehog pathway via PI3K–Akt in human esophageal adenocarcinoma: Identification of novel targets for concerted therapy concepts

Maxim Kebenko; Astrid Drenckhan; Stephanie J. Gros; Manfred Jücker; Nicole Grabinski; Florian Ewald; Astrid Grottke; Alexander Schultze; Jakob R. Izbicki; Carsten Bokemeyer; Jasmin Wellbrock; Walter Fiedler

The Hedgehog pathway plays an important role in the pathogenesis of several tumor types, including esophageal cancer. In our study, we show an expression of the ligand Indian hedgehog (Ihh) and its downstream mediator Gli-1 in primary resected adenocarcinoma tissue by immunohistochemistry and quantitative PCR in fifty percent of the cases, while matching healthy esophagus mucosa was negative for both proteins. Moreover, a functionally important regulation of Gli-1 by ErbB2-PI3K-mTORC signaling as well as a Gli-1-dependent regulation of Ihh in the ErbB2 amplified esophageal adenocarcinoma cell line OE19 was observed. Treatment of OE19 cells with the Her2 antibody trastuzumab, the PI3K-mTORC1 inhibitor NVP BEZ235 (BEZ235) or the knockdown of Akt1 resulted in a downregulation of Gli-1 and Ihh as well as in a reduction of viable OE19 cells in vitro. Interestingly, the Hedgehog receptor Smo, which acts upstream of Gli-1, was not expressed in OE19 cells and in the majority of primary human esophageal adenocarcinoma, suggesting a non-canonical upregulation of Gli-1 expression by the ErbB2-PI3K axis. To translate our findings into a therapeutic concept, we targeted ErbB2-PI3K-mTORC1 by trastuzumab and BEZ235, combining both compounds with the Gli-1/2 inhibitor GANT61. The triple combination led to significantly stronger reduction of tumor cell viability than cisplatinum or each biological alone. Therefore, concomitant blockage of the ErbB2-PI3K pathway and the Hedgehog downstream mediator Gli-1 may provide a new therapeutic strategy for esophageal cancer.


Oncotarget | 2016

Phase I clinical study of RG7356, an anti-CD44 humanized antibody, in patients with acute myeloid leukemia

Norbert Vey; Jacques Delaunay; Giovanni Martinelli; Walter Fiedler; Emmanuel Raffoux; Thomas Prebet; Carlos Gomez-Roca; Cristina Papayannidis; Maxim Kebenko; Peter Paschka; Randolph Christen; Ernesto Guarin; Ann-Marie Bröske; Monika Baehner; Michael Brewster; Antje-Christine Walz; Francesca Michielin; Valeria Runza; Valerie Meresse; Christian Recher

RG7356, a recombinant anti-CD44 immunoglobulin G1 humanized monoclonal antibody, inhibits cell adhesion and has been associated with macrophage activation in preclinical models. We report results of a phase I dose-escalation study of RG7356 in relapsed/refractory acute myeloid leukemia (AML). Eligible patients with refractory AML, relapsed AML after induction chemotherapy, or previously untreated AML not eligible for intensive chemotherapy were enrolled and received intravenous RG7356 at dosages ≤ 2400 mg every other week or ≤ 1200 mg weekly or twice weekly; dose escalation started at 300 mg. Forty-four patients (median age, 69 years) were enrolled. One dose-limiting toxicity occurred (grade 3 hemolysis exacerbation) after one 1200 mg dose (twice-weekly cohort). The majority of adverse events were mild/moderate. Infusion-related reactions occurred in 64% of patients mainly during cycle 1. Two patients experienced grade 3 drug-induced aseptic meningitis. Pharmacokinetics increased supraproportionally, suggesting a target-mediated drug disposition (TMDD) at ≥ 1200 mg. Two patients achieved complete response with incomplete platelet recovery or partial response, respectively. One patient had stable disease with hematologic improvement. RG7356 was generally safe and well tolerated. Maximum tolerated dose was not reached, but saturation of TMDD was achieved. The recommended dose for future AML evaluations is 2400 mg every other week.


Oncotarget | 2017

Combined inhibition of GLI and FLT3 signaling leads to effective anti-leukemic effects in human acute myeloid leukemia

Emily-Marie Latuske; Hauke Stamm; Marianne Klokow; Gabi Vohwinkel; Jana Muschhammer; Carsten Bokemeyer; Manfred Jücker; Maxim Kebenko; Walter Fiedler; Jasmin Wellbrock

Activation of the Hedgehog pathway has been implicated in the pathogenesis of several tumor types including myeloid leukemia. Previously we demonstrated that overexpression of Hedgehog downstream mediators GLI1/2 confers an adverse prognosis to patients with acute myeloid leukemia (AML) and is correlated with a FLT3 mutated status. To analyze a possible non-canonical activation of the Hedgehog pathway via FLT3 and PI3K, we performed blocking experiments utilizing inhibitors for FLT3 (sunitinib), PI3K (PF-04691502) and GLI1/2 (GANT61) in FLT3-mutated and FLT3 wildtype AML cell lines and primary blasts. Combination of all three compounds had stronger anti-leukemic effects in FLT3-mutated compared to FLT3 wildtype AML cells in vitro. Interestingly, the colony growth of normal CD34+ cells from healthy donors was not impeded by the triple inhibitor combination possibly opening a therapeutic window for the clinical use of inhibitor combinations. Besides, combined treatment with sunitinib, PF-04691502 and GANT61 significantly prolonged the survival of mice transplanted with FLT3-mutated MV4-11 cells compared to the single agent treatments. Furthermore, the inhibition of FLT3 and PI3K resulted in reduced GLI protein expression and promotor activity in FLT3-mutated but not in FLT3 wildtype AML cell lines in western blotting and GLI1/2 promoter assays supporting our hypothesis of non-canonical GLI activation via FLT3. In summary, FLT3-mutated in contrast to FLT3 wildtype cells or normal human hematopoietic progenitor cells are exquisitely sensitive to combined inhibition by FLT3, PI3K and GLI1/2 overcoming some of the limitations of current FLT3 directed therapy in AML. The development of GLI1/2 inhibitors is highly desirable.


OncoImmunology | 2018

A multicenter phase 1 study of solitomab (MT110, AMG 110), a bispecific EpCAM/CD3 T-cell engager (BiTE®) antibody construct, in patients with refractory solid tumors

Maxim Kebenko; Marie Elisabeth Goebeler; Martin Wolf; Annette Hasenburg; Ruth Seggewiss-Bernhardt; Barbara Ritter; Beate Rautenberg; Djordje Atanackovic; Andrea Kratzer; James B. Rottman; Matthias Friedrich; Eva Vieser; Stefanie Elm; Ingrid Patzak; Dorothea Wessiepe; Sabine Stienen; Walter Fiedler

ABSTRACT We assessed the tolerability and antitumor activity of solitomab, a bispecific T-cell engager (BiTE®) antibody construct targeting epithelial cell adhesion molecule (EpCAM). Patients with relapsed/refractory solid tumors not amenable to standard therapy received solitomab as continuous IV infusion in a phase 1 dose-escalation study with six different dosing schedules. The primary endpoint was frequency and severity of adverse events (AEs). Secondary endpoints included pharmacokinetics, pharmacodynamics, immunogenicity, and antitumor activity. Sixty-five patients received solitomab at doses between 1 and 96 µg/day for ≥28 days. Fifteen patients had dose-limiting toxicities (DLTs): eight had transient abnormal liver parameters shortly after infusion start or dose escalation (grade 3, n = 4; grade 4, n = 4), and one had supraventricular tachycardia (grade 3); all events resolved with solitomab discontinuation. Six patients had a DLT of diarrhea: four events resolved (grade 3, n = 3; grade 4, n = 1), one (grade 3) was ongoing at the time of treatment-unrelated death, and one (grade 3) progressed to grade 5 after solitomab discontinuation. The maximum tolerated dose was 24 µg/day. Overall, 95% of patients had grade ≥3 treatment-related AEs, primarily diarrhea, elevated liver parameters, and elevated lipase. Solitomab half-life was 4.5 hours; serum levels plateaued within 24 hours. One unconfirmed partial response was observed. In this study of a BiTE® antibody construct targeting solid tumors, treatment of relapsed/refractory EpCAM-positive solid tumors with solitomab was associated with DLTs, including severe diarrhea and increased liver enzymes, which precluded dose escalation to potentially therapeutic levels.


Blood | 2014

Favorable Outcome in a Large Cohort of Prospectively Treated Adult Patients with T-Lymphoblastic Lymphoma (T-LBL) Despite Slowly Evolving Complete Remission Assessed By Conventional Radiography

Nicola Gökbuget; Andrea Wolf; Matthias Stelljes; Andreas Hüttmann; Eike C. Buss; Andreas Viardot; Kalina Brandt; Maike de Wit; Norbert Frickhofen; Maxim Kebenko; Mustafa Kondakci; Doris Kraemer; Stefan Schwartz; Hubert Serve; Karsten Spiekermann; Reingard Stuhlmann; Albrecht Reichle; Dieter Hoelzer


Journal of Clinical Oncology | 2017

A phase I study of EpCAM/CD3-bispecific antibody (MT110) in patients with advanced solid tumors.

Walter Fiedler; Martin Wolf; Maxim Kebenko; Marie-Elisabeth Goebeler; Barbara Ritter; Alexander Quaas; Eva Vieser; Youssef Hijazi; Ingrid Patzak; Matthias G. Friedrich; Peter Kufer; Stanley R. Frankel; Ruth Seggewiss-Bernhardt; Sabine Kaubitzsch


Blood | 2012

Sunitinib and Intensive Chemotherapy in Patients with Acute Myeloid Leukemia and Activating FLT3 Mutations: Results of the AMLSG 10-07 Study (ClinicalTrials.gov No. NCT00783653)

Walter Fiedler; Sabine Kayser; Maxim Kebenko; Jürgen Krauter; Helmut R. Salih; Katharina Götze; Daniela Späth; Gudrun Göhring; Veronica Teleanu; Konstanze Döhner; Arnold Ganser; Hartmut Döhner; Richard F. Schlenk


Journal of Clinical Oncology | 2016

A first-in-patient phase I study of BGB324, a selective Axl kinase inhibitor in patients with refractory/relapsed AML and high-risk MDS.

Sonja Loges; Bjorn Torre Gjertsen; Michael Heuser; Isabel Ben-Batalla; David R. Micklem; Chromik Jorg; Maxim Kebenko; Walter Fiedler; Jorge Cortes


Blood | 2015

Selinexor, ARA-C and Idarubicin: An Effective and Tolerable Combination in Patients with Relapsed/Refractory AML: A Multicenter Phase II Study

Walter Fiedler; Jörg Chromik; Maxim Kebenko; Felicitas Thol; Arne Trummer; Chirstoph Schünemann; Christian Brandts; Anne Köhler; Vera Schlipfenbacher; Carsten Bokemeyer; Susann Theile; Anne L. Kranich; Michael Heuser

Collaboration


Dive into the Maxim Kebenko's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jörg Chromik

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Jorge Cortes

University of Texas MD Anderson Cancer Center

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