Network


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

Hotspot


Dive into the research topics where Roland Neuhaus is active.

Publication


Featured researches published by Roland Neuhaus.


ChemMedChem | 2016

Identification and Optimization of the First Highly Selective GLUT1 Inhibitor BAY‐876

Holger Siebeneicher; Arwed Cleve; Hartmut Rehwinkel; Roland Neuhaus; Iring Heisler; Thomas Müller; Marcus Bauser; Bernd Buchmann

Despite the long‐known fact that the facilitative glucose transporter GLUT1 is one of the key players safeguarding the increase in glucose consumption of many tumor entities even under conditions of normal oxygen supply (known as the Warburg effect), only few endeavors have been undertaken to find a GLUT1‐selective small‐molecule inhibitor. Because other transporters of the GLUT1 family are involved in crucial processes, these transporters should not be addressed by such an inhibitor. A high‐throughput screen against a library of ∼3 million compounds was performed to find a small molecule with this challenging potency and selectivity profile. The N‐(1H‐pyrazol‐4‐yl)quinoline‐4‐carboxamides were identified as an excellent starting point for further compound optimization. After extensive structure–activity relationship explorations, single‐digit nanomolar inhibitors with a selectivity factor of >100 against GLUT2, GLUT3, and GLUT4 were obtained. The most promising compound, BAY‐876 [N4‐[1‐(4‐cyanobenzyl)‐5‐methyl‐3‐(trifluoromethyl)‐1H‐pyrazol‐4‐yl]‐7‐fluoroquinoline‐2,4‐dicarboxamide], showed good metabolic stability in vitro and high oral bioavailability in vivo.


International Journal of Cancer | 2017

BAY 1125976, a selective allosteric AKT1/2 inhibitor exhibits high efficacy on AKT signaling‐dependent tumor growth in mouse models

Oliver Politz; Franziska Siegel; Lars Bärfacker; Ulf Bömer; Andrea Hägebarth; William Scott; Martin Michels; Stuart Ince; Roland Neuhaus; Kirstin Meyer; Amaury Ernesto Fernández-Montalván; Ningshu Liu; Franz von Nussbaum; Dominik Mumberg; Karl Ziegelbauer

The PI3K‐AKT‐mTOR signaling cascade is activated in the majority of human cancers, and its activation also plays a key role in resistance to chemo and targeted therapeutics. In particular, in both breast and prostate cancer, increased AKT pathway activity is associated with cancer progression, treatment resistance and poor disease outcome. Here, we evaluated the activity of a novel allosteric AKT1/2 inhibitor, BAY 1125976, in biochemical, cellular mechanistic, functional and in vivo efficacy studies in a variety of tumor models. In in vitro kinase activity assays, BAY 1125976 potently and selectively inhibited the activity of full‐length AKT1 and AKT2 by binding into an allosteric binding pocket formed by kinase and PH domain. In accordance with this proposed allosteric binding mode, BAY 1125976 bound to inactive AKT1 and inhibited T308 phosphorylation by PDK1, while the activity of truncated AKT proteins lacking the pleckstrin homology domain was not inhibited. In vitro, BAY 1125976 inhibited cell proliferation in a broad panel of human cancer cell lines. Particularly high activity was observed in breast and prostate cancer cell lines expressing estrogen or androgen receptors. Furthermore, BAY 1125976 exhibited strong in vivo efficacy in both cell line and patient‐derived xenograft models such as the KPL4 breast cancer model (PIK3CAH1074R mutant), the MCF7 and HBCx‐2 breast cancer models and the AKTE17K mutant driven prostate cancer (LAPC‐4) and anal cancer (AXF 984) models. These findings indicate that BAY 1125976 is a potent and highly selective allosteric AKT1/2 inhibitor that targets tumors displaying PI3K/AKT/mTOR pathway activation, providing opportunities for the clinical development of new, effective treatments.


bioRxiv | 2018

Targeting a Therapy-Resistant Cancer Cell State Using Masked Electrophiles as GPX4 Inhibitors

John K. Eaton; Laura Furst; Richard A Ruberto; Dieter Moosmayer; Roman Hillig; Andre Hilpmann; Katja Zimmermann; Matthew J. Ryan; Michael Niehues; Volker Badock; Anneke Kramm; Sixun Chen; Paul A. Clemons; Stefan Gradl; Claire Montagnon; Kiel E Lazarski; Sven Christian; Besnik Bajrami; Roland Neuhaus; Ashley L Eheim; Vasanthi Viswanathan; Stuart L. Schreiber

We recently discovered that inhibition of the lipid peroxidase GPX4 can selectively kill cancer cells in a therapy-resistant state through induction of ferroptosis. Although GPX4 lacks a conventional druggable pocket, covalent small-molecule inhibitors are able to overcome this challenge by reacting with the GPX4 catalytic selenocysteine residue to eliminate enzymatic activity. Unfortunately, all currently-reported GPX4 inhibitors achieve their activity through a reactive chloroacetamide group; this dependence hinders their selectivity and stability and makes them unsuitable for use in vivo. Development of therapeutically useful GPX4 inhibitors may be achieved by the identification of new electrophilic chemotypes and mechanisms of action that do not suffer these shortcomings. Here, we report our discovery that nitrile oxide electrophiles, and a set of remarkable chemical transformations that generates them in cells from masked precursors, provide an effective strategy for selective targeting of GPX4. Our results, which include structural insights, target engagement assays, and diverse GPX4-inhibitor tool compounds, provide critical insights that may galvanize development of therapeutic agents for exploring the efficacy and safety of inhibiting the currently-undruggable GPX4. Our discovery that nitrile oxide electrophiles engage in highly selective cellular interactions and are bioavailable in their masked forms may also be valuable in the development of covalent inhibitors of other challenging targets.


Cancer Research | 2016

Abstract 4746: Pharmacological characterization of BAY-876, a novel highly selective inhibitor of glucose transporter (GLUT)-1 in vitro and in vivo

Charlotte Kopitz; Luisella Toschi; Carolyn Algire; Mélanie Héroult; Anna-Lena Frisk; Kirstin Meyer; Arndt Schmitz; Eleni Lagkadinou; Heike Petrul; Iring Heisler; Roland Neuhaus; Bernd Buchmann; Herbert Himmel; Marcus Bauser; Andrea Haegebarth; Karl Ziegelbauer

One hallmark of cancer is the accelerated metabolism, high energy requirements, and increased glucose uptake by the tumor cells, the latter being the first and rate-limiting step for glucose metabolism. Glucose transport into the tumor cell is mediated by facilitative high-affinity glucose transporter (GLUT) proteins. Among the 14 GLUT proteins, expression of GLUT1 in normal organs is nearly exclusively restricted to the blood brain barrier, while other GLUTs are also expressed in a wide variety of vital organs such as liver and heart. Interestingly, GLUT1 expression is highly regulated by hypoxia-inducible factor (HIF)-1α, a key driver of tumor progression. In line with this finding, GLUT1 over-expression was found to be associated with tumor progression and poor overall survival in various tumor indications. Consequently, GLUT1 represents a potential target for cancer treatment. Therefore, we have developed a highly-selective GLUT1 inhibitor, namely BAY-876, with selectivity over GLUT2, 3, and 4 of 4700-, 800-, and 135-fold, respectively. We here show for the first time the pharmacological characterization of BAY-876, comprising inhibition of glucose-uptake, anti-proliferative activity in vitro, and anti-tumor efficacy in vivo in models of different tumor indications in monotherapy as well as first results on the combinability of BAY-876. Furthermore, at the therapeutic dose, BAY-876 treatment did not show any relevant finding on the behavior of treated mice in the Irwin test, assuming no or only minor effects on brain function. In conclusion, BAY-876 is the first GLUT1-selective inhibitor which reduces glucose uptake and growth of tumor cells with sufficient tolerability at the efficacious dose in preclinical models. Citation Format: Charlotte Kopitz, Luisella Toschi, Carolyn Algire, Melanie Heroult, Anna-Lena Frisk, Kirstin Meyer, Arndt Schmitz, Eleni Lagkadinou, Heike Petrul, Iring Heisler, Roland Neuhaus, Bernd Buchmann, Herbert Himmel, Marcus Bauser, Andrea Haegebarth, Karl Ziegelbauer. Pharmacological characterization of BAY-876, a novel highly selective inhibitor of glucose transporter (GLUT)-1 in vitro and in vivo. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4746.


Archive | 2011

Substituted imidazo[1,2-a]pyrimidines and —pyridines

Stuart Ince; Andrea Hägebarth; Oliver Politz; Roland Neuhaus; Ulf Bömer; William J. Scott


Archive | 2013

SUBSTITUTED PYRIMIDO[1,2-B]INDAZOLES AND THEIR USE AS MODULATORS OF THE PI3K/AKT PATHWAY

Hartmut Rehwinkel; Andrea Hägebarth; Oliver Politz; Roland Neuhaus; Ulf Bömer


Archive | 2012

Imidazopyridazines as akt kinase inhibitors

Lars Bärfacker; William Johnston Scott; Andrea Hägebarth; Stuart Ince; Hartmut Rehwinkel; Oliver Politz; Roland Neuhaus; Hans Briem; Ulf Bömer


Archive | 2011

SUBSTITUTED IMIDAZO[1,2-B]PYRIDAZINES

Hartmut Rehwinkel; Andrea Hägebarth; Oliver Politz; Roland Neuhaus; Ulf Bömer


Molecular Cancer Therapeutics | 2018

Preclinical Efficacy of the Novel Monocarboxylate Transporter 1 Inhibitor BAY-8002 and Associated Markers of Resistance

Maria Quanz; Eckhard Bender; Charlotte Kopitz; Sylvia Grünewald; Andreas Schlicker; Wolfgang Schwede; Ashley L Eheim; Luisella Toschi; Roland Neuhaus; Carmen Richter; Joern Toedling; Claudia Merz; Ralf Lesche; Atanas Kamburov; Holger Siebeneicher; Marcus Bauser; Andrea Haegebarth


ChemMedChem | 2016

Cover Picture: Identification and Optimization of the First Highly Selective GLUT1 Inhibitor BAY‐876 (ChemMedChem 20/2016)

Holger Siebeneicher; Arwed Cleve; Hartmut Rehwinkel; Roland Neuhaus; Iring Heisler; Thomas Müller; Marcus Bauser; Bernd Buchmann

Researchain Logo
Decentralizing Knowledge