Network


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

Hotspot


Dive into the research topics where Philip Lienau is active.

Publication


Featured researches published by Philip Lienau.


Neuro-oncology | 2009

Sagopilone crosses the blood-brain barrier in vivo to inhibit brain tumor growth and metastases.

Jens Hoffmann; Iduna Fichtner; Margit Lemm; Philip Lienau; Holger Hess-Stumpp; Andrea Rotgeri; Birte Hofmann; Ulrich Klar

The aim of this study was to determine the efficacy of sagopilone (ZK-EPO), a novel epothilone, compared with other anticancer agents in orthotopic models of human primary and secondary brain tumors. Autoradiography and pharmacokinetic analyses were performed on rats and mice to determine passage across the blood-brain barrier and organ distribution of sagopilone. Mice bearing intracerebral human tumors (U373 or U87 glioblastoma, MDA-MB-435 melanoma, or patient-derived non-small-cell lung cancer [NSCLC]) were treated with sagopilone 5-10 mg/kg, paclitaxel 8-12.5 mg/kg (or temozolomide, 100 mg/kg) or control (vehicle only). Tumor volume was measured to assess antitumor activity. Sagopilone crossed the blood-brain barrier in both rat and mouse models, leading to therapeutically relevant concentrations in the brain with a long half-life. Sagopilone exhibited significant antitumor activity in both the U373 and U87 models of human glioblastoma, while paclitaxel showed a limited effect in the U373 model. Sagopilone significantly inhibited the growth of tumors from CNS metastasis models (MDA-MB-435 melanoma and patient-derived Lu7187 and Lu7466 NSCLC) implanted in the brains of nude mice, in contrast to paclitaxel or temozolomide. Sagopilone has free access to the brain. Sagopilone demonstrated significant antitumor activity in orthotopic models of both glioblastoma and CNS metastases compared with paclitaxel or temozolomide, underlining the value of further research evaluating sagopilone in the treatment of brain tumors. Sagopilone is currently being investigated in a broad phase II clinical trial program, including patients with glioblastoma, NSCLC, breast cancer, and melanoma.


Cell Death & Differentiation | 2013

Characterization of novel MPS1 inhibitors with preclinical anticancer activity

M Jemaà; Lorenzo Galluzzi; Oliver Kepp; Laura Senovilla; M Brands; U Boemer; M Koppitz; Philip Lienau; S Prechtl; V Schulze; Gerhard Siemeister; A M Wengner; D Mumberg; K Ziegelbauer; A Abrieu; Maria Castedo; Ilio Vitale; Guido Kroemer

Monopolar spindle 1 (MPS1), a mitotic kinase that is overexpressed in several human cancers, contributes to the alignment of chromosomes to the metaphase plate as well as to the execution of the spindle assembly checkpoint (SAC). Here, we report the identification and functional characterization of three novel inhibitors of MPS1 of two independent structural classes, N-(4-{2-[(2-cyanophenyl)amino][1,2,4]triazolo[1,5-a]pyridin-6-yl}phenyl)-2-phenylacetamide (Mps-BAY1) (a triazolopyridine), N-cyclopropyl-4-{8-[(2-methylpropyl)amino]-6-(quinolin-5-yl)imidazo[1,2-a]pyrazin-3-yl}benzamide (Mps-BAY2a) and N-cyclopropyl-4-{8-(isobutylamino)imidazo[1,2-a]pyrazin-3-yl}benzamide (Mps-BAY2b) (two imidazopyrazines). By selectively inactivating MPS1, these small inhibitors can arrest the proliferation of cancer cells, causing their polyploidization and/or their demise. Cancer cells treated with Mps-BAY1 or Mps-BAY2a manifested multiple signs of mitotic perturbation including inefficient chromosomal congression during metaphase, unscheduled SAC inactivation and severe anaphase defects. Videomicroscopic cell fate profiling of histone 2B-green fluorescent protein-expressing cells revealed the capacity of MPS1 inhibitors to subvert the correct timing of mitosis as they induce a premature anaphase entry in the context of misaligned metaphase plates. Hence, in the presence of MPS1 inhibitors, cells either divided in a bipolar (but often asymmetric) manner or entered one or more rounds of abortive mitoses, generating gross aneuploidy and polyploidy, respectively. In both cases, cells ultimately succumbed to the mitotic catastrophe-induced activation of the mitochondrial pathway of apoptosis. Of note, low doses of MPS1 inhibitors and paclitaxel (a microtubular poison) synergized at increasing the frequency of chromosome misalignments and missegregations in the context of SAC inactivation. This resulted in massive polyploidization followed by the activation of mitotic catastrophe. A synergistic interaction between paclitaxel and MPS1 inhibitors could also be demonstrated in vivo, as the combination of these agents efficiently reduced the growth of tumor xenografts and exerted superior antineoplastic effects compared with either compound employed alone. Altogether, these results suggest that MPS1 inhibitors may exert robust anticancer activity, either as standalone therapeutic interventions or combined with microtubule-targeting chemicals.


ACS Chemical Biology | 2016

Conformational Adaption May Explain the Slow Dissociation Kinetics of Roniciclib (BAY 1000394), a Type I CDK Inhibitor with Kinetic Selectivity for CDK2 and CDK9.

Pelin Ayaz; Dorothee Andres; Dennis Kwiatkowski; Carl-Christian Kolbe; Philip Lienau; Gerhard Siemeister; Ulrich Lücking; Christian Stegmann

Roniciclib (BAY 1000394) is a type I pan-CDK (cyclin-dependent kinase) inhibitor which has revealed potent efficacy in xenograft cancer models. Here, we show that roniciclib displays prolonged residence times on CDK2 and CDK9, whereas residence times on other CDKs are transient, thus giving rise to a kinetic selectivity of roniciclib. Surprisingly, variation of the substituent at the 5-position of the pyrimidine scaffold results in changes of up to 3 orders of magnitude of the drug-target residence time. CDK2 X-ray cocrystal structures have revealed a DFG-loop adaption for the 5-(trifluoromethyl) substituent, while for hydrogen and bromo substituents the DFG loop remains in its characteristic type I inhibitor position. In tumor cells, the prolonged residence times of roniciclib on CDK2 and CDK9 are reflected in a sustained inhibitory effect on retinoblastoma protein (RB) phosphorylation, indicating that the target residence time on CDK2 may contribute to sustained target engagement and antitumor efficacy.


Blood | 2017

Cyclin-dependent kinase 9 is a novel specific molecular target in adult T-cell leukemia/lymphoma

Tomoko Narita; Takashi Ishida; Asahi Ito; Ayako Masaki; Shiori Kinoshita; Susumu Suzuki; Hisashi Takino; Takashi Yoshida; Masaki Ri; Shigeru Kusumoto; Hirokazu Komatsu; Kazunori Imada; Yuetsu Tanaka; Akifumi Takaori-Kondo; Hiroshi Inagaki; Arne Scholz; Philip Lienau; Taruho Kuroda; Ryuzo Ueda; Shinsuke Iida

Cyclin-dependent kinase 9 (CDK9), a subunit of the positive transcription elongation factor b (P-TEFb) complex, regulates gene transcription elongation by phosphorylating the C-terminal domain (CTD) of RNA polymerase II (RNAPII). The deregulation of CDK9/P-TEFb has important implications for many cancer types. BAY 1143572 is a novel and highly selective CDK9/P-TEFb inhibitor currently being investigated in phase 1 studies. We evaluated the therapeutic potential of BAY 1143572 in adult T-cell leukemia/lymphoma (ATL). As a result of CDK9 inhibition and subsequent inhibition of phosphorylation at serine 2 of the RNAPII CTD, BAY 1143572 decreased c-Myc and Mcl-1 levels in ATL-derived or human T-cell lymphotropic virus type-1 (HTLV-1)-transformed lines and primary ATL cells tested, leading to their growth inhibition and apoptosis. Median inhibitory concentrations for BAY 1143572 in ATL-derived or HTLV-1-transformed lines (n = 8), primary ATL cells (n = 11), and CD4+ cells from healthy volunteers (n = 5) were 0.535, 0.30, and 0.36 μM, respectively. Next, NOG mice were used as recipients of tumor cells from an ATL patient. BAY 1143572-treated ATL-bearing mice (once daily 12.5 mg/kg oral application) demonstrated significantly decreased ATL cell infiltration of the liver and bone marrow, as well as decreased human soluble interleukin-2 receptor levels in serum (reflecting the ATL tumor burden), compared with untreated mice (n = 8 for both). BAY 1143572-treated ATL-bearing mice demonstrated significantly prolonged survival compared with untreated ATL-bearing mice (n = 7 for both). Collectively, this study indicates that BAY 1143572 showed strong potential as a novel treatment of ATL.


ChemMedChem | 2017

Identification of Atuveciclib (BAY 1143572), the First Highly Selective, Clinical PTEFb/CDK9 Inhibitor for the Treatment of Cancer

Ulrich Lücking; Arne Scholz; Philip Lienau; Gerhard Siemeister; Dirk Kosemund; Rolf Bohlmann; Hans Briem; Ildiko Terebesi; Kirstin Meyer; Katja Prelle; Karsten Dr Denner; Ulf Bömer; Martina Schäfer; Knut Eis; Ray Valencia; Stuart Ince; Franz von Nussbaum; Dominik Mumberg; Karl Ziegelbauer; Bert Klebl; Axel Choidas; Peter Nussbaumer; Matthias Baumann; Carsten Schultz-Fademrecht; Gerd Rühter; Jan Eickhoff; Michael Brands

Selective inhibition of exclusively transcription‐regulating PTEFb/CDK9 is a promising new approach in cancer therapy. Starting from lead compound BAY‐958, lead optimization efforts strictly focusing on kinase selectivity, physicochemical and DMPK properties finally led to the identification of the orally available clinical candidate atuveciclib (BAYu20051143572). Structurally characterized by an unusual benzyl sulfoximine group, BAYu20051143572 exhibited the best overall profile inu2005vitro and inu2005vivo, including high efficacy and good tolerability in xenograft models in mice and rats. BAYu20051143572 is the first potent and highly selective PTEFb/CDK9 inhibitor to enter clinical trials for the treatment of cancer.


Cancer Letters | 2017

BAY 1143269, a novel MNK1 inhibitor, targets oncogenic protein expression and shows potent anti-tumor activity

Susann Santag; Franziska Siegel; Antje M. Wengner; Claudia Lange; Ulf Bömer; Knut Eis; Florian Pühler; Philip Lienau; Linda Bergemann; Martin Michels; Franz von Nussbaum; Dominik Mumberg; Kirstin Petersen

The initiation of mRNA translation has received increasing attention as an attractive target for cancer treatment in the recent years. The oncogenic eukaryotic translation initiation factor 4E (eIF4E) is the major substrate of MAP kinase-interacting kinase 1 (MNK1), and it is located at the junction of the cancer-associated PI3K and MAPK pathways. The fact that MNK1 is linked to cell transformation and tumorigenesis renders the kinase a promising target for cancer therapy. We identified a novel small molecule MNK1 inhibitor, BAY 1143269, by high-throughput screening and lead optimization. In kinase assays, BAY 1143269 showed potent and selective inhibition of MNK1. By targeting MNK1 activity, BAYxa01143269 strongly regulated downstream factors involved in cell cycle regulation, apoptosis, immune response and epithelial-mesenchymal transition inxa0vitro or inxa0vivo. In addition, BAY 1143269 demonstrated strong efficacy in monotherapy in cell line and patient-derived non-small cell lung cancer xenograft models as well as delayed tumor regrowth in combination treatment with standard of care chemotherapeutics. In summary, the inhibition of MNK1 activity with a highly potent and selective inhibitor BAY 1143269 may provide an innovative approach for anti-cancer therapy.


Chemistry: A European Journal | 2018

Exploration of Novel Chemical Space: Synthesis and in vitro Evaluation of N-Functionalized Tertiary Sulfonimidamides

Flavia Izzo; Martina Schäfer; Philip Lienau; Ursula Ganzer; Robert A. Stockman; Ulrich Lücking

Abstract An unprecedented set of structurally diverse sulfonimidamides (47u2005compounds) has been prepared by various N‐functionalization reactions of tertiary =NH sulfonimidamide 2u2009aa. These N‐functionalization reactions of model compoundu20052u2009aa include arylation, alkylation, trifluoromethylation, cyanation, sulfonylation, alkoxycarbonylation (carbamate formation) and aminocarbonylation (urea formation). Small molecule X‐ray analyses of selected N‐functionalized products are reported. To gain further insight into the properties of sulfonimidamides relevant to medicinal chemistry, a variety of structurally diverse reaction products were tested in selected in vitro assays. The described N‐functionalization reactions provide a short and efficient approach to structurally diverse sulfonimidamides which have been the subject of recent, growing interest in the life sciences.


Archive | 2012

6H-THIENO[3,2-f][1,2,4]TRIAZOLO[4,3-a][1,4]DIAZEPINE

Norbert Schmees; Joachim Kuhnke; Bernard Haendler; Philip Lienau; Amaury Ernesto Fernandez-Montalvan; Pascale Lejeune; Stephan Siegel; William J. Scott


Archive | 2013

Amino-substituted imidazopyridazines

Knut Eis; Florian Pühler; Ludwig Zorn; Volker Dr Schulze; Detlev Sülzle; Philip Lienau; Andrea Hägebarth; Kirstin Petersen; Ulf Bömer


Archive | 2011

6-SUBSTITUTED IMIDAZOPYRAZINES FOR USE AS MPS-1 AND TKK INHIBITORS IN THE TREATMENT OF HYPERPROLIFERATIVE DISORDERS

Marcus Koppitz; Ulrich Klar; Rolf Jautelat; Dirk Kosemund; Rolf Bohlmann; Benjamin Bader; Philip Lienau; Gerhard Siemeister

Collaboration


Dive into the Philip Lienau's collaboration.

Researchain Logo
Decentralizing Knowledge