Network


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

Hotspot


Dive into the research topics where Rogier C. Buijsman is active.

Publication


Featured researches published by Rogier C. Buijsman.


British Journal of Pharmacology | 2012

A guide to picking the most selective kinase inhibitor tool compounds for pharmacological validation of drug targets

Joost C.M. Uitdehaag; Folkert Verkaar; Husam Alwan; Jos de Man; Rogier C. Buijsman; G.J.R. Zaman

To establish the druggability of a target, genetic validation needs to be supplemented with pharmacological validation. Pharmacological studies, especially in the kinase field, are hampered by the fact that many reference inhibitors are not fully selective for one target. Fortunately, the initial trickle of selective inhibitors released in the public domain has steadily swelled into a stream. However, rationally picking the most selective tool compound out of the increasing amounts of available inhibitors has become progressively difficult due to the lack of accurate quantitative descriptors of drug selectivity. A recently published approach, termed ‘selectivity entropy’, is an improved way of expressing selectivity as a single‐value parameter and enables rank ordering of inhibitors. We provide a guide to select the best tool compounds for pharmacological validation experiments of candidate drug targets using selectivity entropy. In addition, we recommend which inhibitors to use for studying the biology of the 20 most investigated kinases that are clinically relevant: Abl (ABL1), AKT1, ALK, Aurora A/B, CDKs, MET, CSF1R (FMS), EGFR, FLT3, ERBB2 (HER2), IKBKB (IKK2), JAK2/3, JNK1/2/3 (MAPK8/9/10), MEK1/2, PLK1, PI3Ks, p38α (MAPK14), BRAF, SRC and VEGFR2 (KDR).


PLOS ONE | 2014

Comparison of the Cancer Gene Targeting and Biochemical Selectivities of All Targeted Kinase Inhibitors Approved for Clinical Use

Joost C.M. Uitdehaag; Jeroen A.D.M. de Roos; Antoon M. van Doornmalen; Martine B.W. Prinsen; Jos de Man; Yoshinori Tanizawa; Yusuke Kawase; Kohichiro Yoshino; Rogier C. Buijsman; Guido J.R. Zaman

The anti-proliferative activities of all twenty-five targeted kinase inhibitor drugs that are in clinical use were measured in two large assay panels: (1) a panel of proliferation assays of forty-four human cancer cell lines from diverse tumour tissue origins; and (2) a panel of more than 300 kinase enzyme activity assays. This study provides a head-on comparison of all kinase inhibitor drugs in use (status Nov. 2013), and for six of these drugs, the first kinome profiling data in the public domain. Correlation of drug activities with cancer gene mutations revealed novel drug sensitivity markers, suggesting that cancers dependent on mutant CTNNB1 will respond to trametinib and other MEK inhibitors, and cancers dependent on SMAD4 to small molecule EGFR inhibitor drugs. Comparison of cellular targeting efficacies reveals the most targeted inhibitors for EGFR, ABL1 and BRAF(V600E)-driven cell growth, and demonstrates that the best targeted agents combine high biochemical potency with good selectivity. For ABL1 inhibitors, we computationally deduce optimized kinase profiles for use in a next generation of drugs. Our study shows the power of combining biochemical and cellular profiling data in the evaluation of kinase inhibitor drug action.


Annals of Oncology | 2015

Inhibition of the spindle assembly checkpoint kinase TTK enhances the efficacy of docetaxel in a triple-negative breast cancer model.

A.R.R. Maia; J. De Man; U. Boon; A. Janssen; J. Y. Song; Manja Omerzu; Jan Gerard Sterrenburg; M.B.W. Prinsen; Nicole Willemsen-Seegers; J.A.D.M. De Roos; A.M. Van Doornmalen; Joost C.M. Uitdehaag; Geert J. P. L. Kops; J. Jonkers; Rogier C. Buijsman; G.J.R. Zaman; René H. Medema

BACKGROUNDnTriple-negative breast cancers (TNBC) are considered the most aggressive type of breast cancer, for which no targeted therapy exists at the moment. These tumors are characterized by having a high degree of chromosome instability and often overexpress the spindle assembly checkpoint kinase TTK. To explore the potential of TTK inhibition as a targeted therapy in TNBC, we developed a highly potent and selective small molecule inhibitor of TTK, NTRC 0066-0.nnnRESULTS AND CONCLUSIONSnThe compound is characterized by long residence time on the target and inhibits the proliferation of a wide variety of human cancer cell lines with potency in the same range as marketed cytotoxic agents. In cell lines and in mice, NTRC 0066-0 inhibits the phosphorylation of a TTK substrate and induces chromosome missegregation. NTRC 0066-0 inhibits tumor growth in MDA-MB-231 xenografts as a single agent after oral application. To address the effect of the inhibitor in breast cancer, we used a well-defined mouse model that spontaneously develops breast tumors that share key morphologic and molecular features with human TNBC. Our studies show that combination of NTRC 0066-0 with a therapeutic dose of docetaxel resulted in doubling of mouse survival and extended tumor remission, without toxicity. Furthermore, we observed that treatment efficacy is only achieved upon co-administration of the two compounds, which suggests a synergistic in vivo effect. Therefore, we propose TTK inhibition as a novel therapeutic target for neoadjuvant therapy in TNBC.


PLOS Medicine | 2016

IL-7 Receptor Mutations and Steroid Resistance in Pediatric T cell Acute Lymphoblastic Leukemia: A Genome Sequencing Study

Yunlei Li; Jessica Buijs-Gladdines; Kirsten Canté-Barrett; Andrew Stubbs; Eric Vroegindeweij; Willem K. Smits; Ronald van Marion; Winand N. M. Dinjens; Martin A. Horstmann; Roland P. Kuiper; Rogier C. Buijsman; Guido J.R. Zaman; Peter J. van der Spek; Rob Pieters; Jules P.P. Meijerink

Background Pediatric acute lymphoblastic leukemia (ALL) is the most common childhood cancer and the leading cause of cancer-related mortality in children. T cell ALL (T-ALL) represents about 15% of pediatric ALL cases and is considered a high-risk disease. T-ALL is often associated with resistance to treatment, including steroids, which are currently the cornerstone for treating ALL; moreover, initial steroid response strongly predicts survival and cure. However, the cellular mechanisms underlying steroid resistance in T-ALL patients are poorly understood. In this study, we combined various genomic datasets in order to identify candidate genetic mechanisms underlying steroid resistance in children undergoing T-ALL treatment. Methods and Findings We performed whole genome sequencing on paired pre-treatment (diagnostic) and post-treatment (remission) samples from 13 patients, and targeted exome sequencing of pre-treatment samples from 69 additional T-ALL patients. We then integrated mutation data with copy number data for 151 mutated genes, and this integrated dataset was tested for associations of mutations with clinical outcomes and in vitro drug response. Our analysis revealed that mutations in JAK1 and KRAS, two genes encoding components of the interleukin 7 receptor (IL7R) signaling pathway, were associated with steroid resistance and poor outcome. We then sequenced JAK1, KRAS, and other genes in this pathway, including IL7R, JAK3, NF1, NRAS, and AKT, in these 69 T-ALL patients and a further 77 T-ALL patients. We identified mutations in 32% (47/146) of patients, the majority of whom had a specific T-ALL subtype (early thymic progenitor ALL or TLX). Based on the outcomes of these patients and their prednisolone responsiveness measured in vitro, we then confirmed that these mutations were associated with both steroid resistance and poor outcome. To explore how these mutations in IL7R signaling pathway genes cause steroid resistance and subsequent poor outcome, we expressed wild-type and mutant IL7R signaling molecules in two steroid-sensitive T-ALL cell lines (SUPT1 and P12 Ichikawa cells) using inducible lentiviral expression constructs. We found that expressing mutant IL7R, JAK1, or NRAS, or wild-type NRAS or AKT, specifically induced steroid resistance without affecting sensitivity to vincristine or L-asparaginase. In contrast, wild-type IL7R, JAK1, and JAK3, as well as mutant JAK3 and mutant AKT, had no effect. We then performed a functional study to examine the mechanisms underlying steroid resistance and found that, rather than changing the steroid receptor’s ability to activate downstream targets, steroid resistance was associated with strong activation of MEK-ERK and AKT, downstream components of the IL7R signaling pathway, thereby inducing a robust antiapoptotic response by upregulating MCL1 and BCLXL expression. Both the MEK-ERK and AKT pathways also inactivate BIM, an essential molecule for steroid-induced cell death, and inhibit GSK3B, an important regulator of proapoptotic BIM. Importantly, treating our cell lines with IL7R signaling inhibitors restored steroid sensitivity. To address clinical relevance, we treated primary T-ALL cells obtained from 11 patients with steroids either alone or in combination with IL7R signaling inhibitors; we found that including a MEK, AKT, mTOR, or dual PI3K/mTOR inhibitor strongly increased steroid-induced cell death. Therefore, combining these inhibitors with steroid treatment may enhance steroid sensitivity in patients with ALL. The main limitation of our study was the modest cohort size, owing to the very low incidence of T-ALL. Conclusions Using an unbiased sequencing approach, we found that specific mutations in IL7R signaling molecules underlie steroid resistance in T-ALL. Future prospective clinical studies should test the ability of inhibitors of MEK, AKT, mTOR, or PI3K/mTOR to restore or enhance steroid sensitivity and improve clinical outcome.


Journal of Biomolecular Screening | 2014

High-throughput fluorescence-based screening assays for tryptophan-catabolizing enzymes.

Nicole Seegers; Antoon M. van Doornmalen; Joost C.M. Uitdehaag; Jos de Man; Rogier C. Buijsman; Guido J.R. Zaman

Indoleamine 2,3-dioxygenase (IDO1) and tryptophan 2,3-dioxygenase (TDO) are two structurally different enzymes that have a different tissue distribution and physiological roles, but both catalyze the conversion of tryptophan to N-formylkynurenine (NFK). IDO1 has been clinically validated as a small-molecule drug target for cancer, while preclinical studies indicate that TDO may be a target for cancer immunotherapy and neurodegenerative disease. We have developed a high-throughput screening assay for IDO1 and TDO based on a novel chemical probe, NFK Green, that reacts specifically with NFK to form a green fluorescent molecule with an excitation wavelength of 400 nm and an emission wavelength of 510 nm. We provide the first side-by-side comparison of a number of published inhibitors of IDO1 and TDO and reveal that the preclinical IDO1 inhibitor Compound 5l shows significant cross-reactivity with TDO, while the relative selectivity of other published inhibitors was confirmed. The suitability for high-throughput screening of the assays was demonstrated by screening a library of 87,000 chemical substances in 384- or 1536-well format. Finally, we demonstrate that the assay can also be used to measure the capacity of cells to metabolize tryptophan and to measure the cellular potency of IDO1 and TDO inhibitors.


PLOS ONE | 2015

Selective Targeting of CTNNB1- , KRAS- or MYC- Driven Cell Growth by Combinations of Existing Drugs

Joost C.M. Uitdehaag; Jeroen A.D.M. de Roos; Antoon M. van Doornmalen; Martine B.W. Prinsen; Jill A. P. Spijkers-Hagelstein; Judith R.F. de Vetter; Jos de Man; Rogier C. Buijsman; Guido J.R. Zaman

The aim of combination drug treatment in cancer therapy is to improve response rate and to decrease the probability of the development of drug resistance. Preferably, drug combinations are synergistic rather than additive, and, ideally, drug combinations work synergistically only in cancer cells and not in non-malignant cells. We have developed a workflow to identify such targeted synergies, and applied this approach to selectively inhibit the proliferation of cell lines with mutations in genes that are difficult to modulate with small molecules. The approach is based on curve shift analysis, which we demonstrate is a more robust method of determining synergy than combination matrix screening with Bliss-scoring. We show that the MEK inhibitor trametinib is more synergistic in combination with the BRAF inhibitor dabrafenib than with vemurafenib, another BRAF inhibitor. In addition, we show that the combination of MEK and BRAF inhibitors is synergistic in BRAF-mutant melanoma cells, and additive or antagonistic in, respectively, BRAF-wild type melanoma cells and non-malignant fibroblasts. This combination exemplifies that synergistic action of drugs can depend on cancer genotype. Next, we used curve shift analysis to identify new drug combinations that specifically inhibit cancer cell proliferation driven by difficult-to-drug cancer genes. Combination studies were performed with compounds that as single agents showed preference for inhibition of cancer cells with mutations in either the CTNNB1 gene (coding for β-catenin), KRAS, or cancer cells expressing increased copy numbers of MYC. We demonstrate that the Wnt-pathway inhibitor ICG-001 and trametinib acted synergistically in Wnt-pathway-mutant cell lines. The ERBB2 inhibitor TAK-165 was synergistic with trametinib in KRAS-mutant cell lines. The EGFR/ERBB2 inhibitor neratinib acted synergistically with the spindle poison docetaxel and with the Aurora kinase inhibitor GSK-1070916 in cell lines with MYC amplification. Our approach can therefore efficiently discover novel drug combinations that selectively target cancer genes.


Angewandte Chemie | 2017

Optimized Target Residence Time: Type I1/2 Inhibitors for p38α MAP Kinase with Improved Binding Kinetics through Direct Interaction with the R-Spine

Heike K. Wentsch; Niklas M. Walter; Mike Bührmann; Svenja Mayer-Wrangowski; Daniel Rauh; Guido J.R. Zaman; Nicole Willemsen-Seegers; Rogier C. Buijsman; Melanie Henning; Daniel Dauch; Lars Zender; Stefan Laufer

Skepinone-L was recently reported to be a p38α MAP kinase inhibitor with high potency and excellent selectivity inu2005vitro and inu2005vivo. However, this class of compounds still act as fully ATP-competitive Typeu2005I binders which, furthermore, suffer from short residence times at the enzyme. We herein describe a further development with the first Typeu2005I1/2 binders for p38α MAP kinase. Typeu2005I1/2 inhibitors interfere with the R-spine, inducing a glycine flip and occupying both hydrophobic regionsu2005I and II. This design approach leads to prolonged target residence time, binding to both the active and inactive states of the kinase, excellent selectivity, excellent potency on the enzyme level, and low nanomolar activity in a human whole blood assay. This promising binding mode is proven by X-ray crystallography.


Oncotarget | 2017

Stable aneuploid tumors cells are more sensitive to TTK inhibition than chromosomally unstable cell lines

Marion A.A. Libouban; Jeroen A.D.M. de Roos; Joost C.M. Uitdehaag; Nicole Willemsen-Seegers; Sara Mainardi; Jelle Dylus; Jos de Man; Bastiaan Tops; Jules P.P. Meijerink; Zuzana Storchova; Rogier C. Buijsman; René H. Medema; Guido J.R. Zaman

Inhibition of the spindle assembly checkpoint kinase TTK causes chromosome mis-segregation and tumor cell death. However, high levels of TTK correlate with chromosomal instability (CIN), which can lead to aneuploidy. We show that treatment of tumor cells with the selective small molecule TTK inhibitor NTRC 0066-0 overrides the mitotic checkpoint, irrespective of cell line sensitivity. In stable aneuploid cells NTRC 0066-0 induced acute CIN, whereas in cells with high levels of pre-existing CIN there was only a small additional fraction of cells mis-segregating their chromosomes. In proliferation assays stable aneuploid cells were more sensitive than cell lines with pre-existing CIN. Tetraploids are thought to be an intermediate between diploid and unstable aneuploid cells. TTK inhibitors had the same potency on post-tetraploid and parental diploid cells, which is remarkable because the post-tetraploids are more resistant to mitotic drugs. Finally, we confirm that the reference compound reversine is a TTK inhibitor and like NTRC 0066-0, inhibits the proliferation of patient-derived colorectal cancer organoids. In contrast, treatment with TTK inhibitor did not reduce the viability of non-proliferating T cell acute lymphoblastic leukemia cells samples. Consequently, TTK inhibitor therapy is expected to spare non-dividing cells, and may be used to target stable aneuploid tumors.


Journal of Medicinal Chemistry | 2017

Design, Synthesis, and Biological Evaluation of Novel Type I1/2 p38α MAP Kinase Inhibitors with Excellent Selectivity, High Potency, and Prolonged Target Residence Time by Interfering with the R-Spine

Niklas M. Walter; Heike K. Wentsch; Mike Bührmann; Silke M. Bauer; Eva Döring; Svenja Mayer-Wrangowski; Adrian Sievers-Engler; Nicole Willemsen-Seegers; Guido J.R. Zaman; Rogier C. Buijsman; Michael Lämmerhofer; Daniel Rauh; Stefan Laufer

We recently reported 1a (skepinone-L) as a type I p38α MAP kinase inhibitor with high potency and excellent selectivity in vitro and in vivo. However, as a type I inhibitor, it is entirely ATP-competitive and shows just a moderate residence time. Thus, the scope was to develop a new class of advanced compounds maintaining the structural binding features of skepinone-L scaffold like inducing a glycine flip at the hinge region and occupying both hydrophobic regions I and II. Extending this scaffold with suitable residues resulted in an interference with the kinases R-Spine. By synthesizing 69 compounds, we could significantly prolong the target residence time with one example to 3663 s, along with an excellent selectivity score of 0.006 and an outstanding potency of 1.0 nM. This new binding mode was validated by cocrystallization, showing all binding interactions typifying type I1/2 binding. Moreover, microsomal studies showed convenient metabolic stability of the most potent, herein reported representatives.


Poster Presentation: Experimental/Molecular Therapeutics, Pharmacogenomics | 2018

PO-484 Development of a two-step screening-and-confirmation approach to efficiently identify synergistic drug combinations with the PARP inhibitor niraparib

M.B.W. Prinsen; Jrf De Vetter; Jelle Dylus; JADMDe Roos; Sjc Van Gerwen; Rogier C. Buijsman; Jcm Uitdehaag; G.J.R. Zaman

Introduction Poly (ADP-ribose) polymerases (PARPs) are important players in DNA damage repair. Inhibition of PARP activity is highly effective against cancers deficient in homologous recombination repair due to, a.o., BRCA1/2 mutations. Clinically, PARP inhibitors (PARPi) are used to treat, a.o., ovarium cancers. Although effective, repeated treatment with PARPi may lead to resistance. To identify new synergistic drug combinations, we developed a two-step approach of screening and confirmation using two of our platform technologies SynergyScreen and SynergyFinder. We first screen for synergy in presence of a fixed concentration of compound (e.g., PARPi), followed by confirmation using dose response curves. The PARPi niraparib was used as a proof-of-concept combinatorial drug at a fixed concentration, and combined with over 150 anti-cancer agents. Material and methods Proliferation of cell lines and patient derived primary ovarium cancer cells was measured using ATPLite 1 Step after 120u2009hours compound incubation. Growth was calculated relative to vehicle treated cells; relative IC50s were calculated from a four parameter logistics curve. The fixed concentration niraparib used in the SynergyScreen represented 80% viability. Anti-cancer agents were screened in absence and presence of niraparib, followed by analysis of IC50 shifts. Combinations with IC50shifts>2u2009fold indicated a synergistic hit and were re-examined using equipotent mixtures and calculation of Combination Index (CI) within our SynergyFinder platform.1 CI <1 indicates synergy. As a control, the combinatorial drug was tested against itself. Results and discussions Results of the SynergyScreen showed various synergistic drug combinations from the high-throughput setup, i.e. niraparib in combination with the topoisomerase I inhibitor irinotecan (CI0.5=0.70) and the DNA alkylating agent temozolomide (CI0.5=0.42). These and other synergies were confirmed in patient samples. Independently from niraparib, the bromodomain inhibitor JQ1 and the histone acetyltransferase inhibitor anacardic acid showed synergy (CI0.5 0.42), which was not demonstrated before. Conclusion Our Synergy platforms can identify novel and reproducible synergistic drug combinations in an unbiased and efficient manner. Proof-of-concept was demonstrated and new synergistic drug combinations were found. Our method is therefore excellently suited for the in vitro discovery and validation of synergistic combinations. Reference Uitdehaag, et al. PLoS ONE 2015;10(5): e0125021.

Collaboration


Dive into the Rogier C. Buijsman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

René H. Medema

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Rauh

Technical University of Dortmund

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge