Publications by Oncolines Scientists

  • Jang et al. (2023) Comparative biochemical kinase activity analysis identifies rivoceranib as a highly selective VEGFR2 inhibitor. Cancer Chemotherapy and Pharmacology, 91:491–499. (Collaboration with Elevar Therapeutics)
  • Grobben et al. (2023) Amino acid-metabolizing enzymes in advanced high-grade serous ovarian cancer patients: value of ascites as biomarker source and role for IL4I1 and IDO1. Cancers, 15(3):893. (Collaboration with Radboud University Medical Center and Pangaea Oncology)
  • Kooijman et al. (2022) Comparative kinase and cancer cell panel profiling of kinase inhibitors approved for clinical use from 2018 to 2020. Frontiers in Oncology, 12:953013.
  • Conlon et al. (2021) Comparative analysis of drug response and gene profiling of the HER2-targeted tyrosine kinase inhibitors. British Journal of Cancer, 124(7):1249–1259. (Collaboration with Dublin City University and Puma Biotechnology, Inc.)
  • Perez-Pardo et al. (2021) Pharmacological validation of TDO as a target for Parkinson’s disease. The FEBS Journal, 288:4311–4331. (Collaboration with Dublin City University and Puma Biotechnology, Inc.)
  • Grobben et al. (2021) Targeting indoleamine 2,3-dioxygenase in cancer models using the novel small molecule inhibitor NTRC 3883-0. Frontiers in Immunology, 11:609490. (Collaboration with Radboud University Medical Center)
  • den Ouden et al. (2020) Chemotherapy sensitivity testing on ovarian cancer cells isolated from malignant ascites. Oncotarget, 11:4570–4581. (Collaboration with Radboud University Medical Center)
  • Grobben et al. (2020) High-throughput fluorescence-based activity assay for Arginase-1. SLAS Discovery, 25(9):1018–1025.
  • Grobben et al. (2020) Structural insights into human Arginase-1 pH dependence and its inhibition by the small molecule inhibitor CB-1158. Journal of Structural Biology: X, 4:100014.
  • Uitdehaag et al. (2019) Combined cellular and biochemical profiling to identify predictive drug response biomarkers for kinase inhibitors approved for clinical use between 2013 and 2017. Molecular Cancer Therapeutics, 18(2):470–481.
  • Zaman et al. (2017) TTK inhibitors as a targeted therapy for CTNNB1 (β-catenin) mutant cancers. Molecular Cancer Therapeutics, 16(11):2609–2617.
  • Libouban et al. (2017) Stable aneuploid tumors cells are more sensitive to TTK inhibition than chromosomally unstable cell lines. Oncotarget, 8(24):38309–38325. (Collaboration with Netherlands Cancer Institute)
  • Uitdehaag et al. (2016) Cell panel profiling reveals conserved therapeutic clusters and differentiates the mechanism of action of different PI3K/mTOR, aurora kinase and EZH2 inhibitors. Molecular Cancer Therapeutics, 15 (12):3097–3109.
  • de Roos JA, Uitdehaag JC, de Man AP, Buijsman RC, Zaman GJR, inventors; Netherlands Translational Research Center B.V., assignee. Prognostic biomarkers for TTK inhibitor chemotherapy. Patent WO 2016/166255 A1. 2016 Oct 20
  • Uitdehaag et al. (2015) Selective targeting of CTNNB1-, KRAS- or MYC-driven cell growth by combinations of existing drugs. PLoS ONE, 10(5):e0125021.
  • Seegers et al. (2014) High-throughput fluorescence-based screening assays for tryptophan-catabolizing enzymes. Journal of Biomolecular Screening, 19(9):1266–1274.
  • Uitdehaag et al. (2014) Comparison of the cancer gene targeting and biochemical selectivities of all targeted kinase inhibitors approved for clinical use. PLoS ONE, 9(3):e92146.

References by Clients to our Services

  • Khameneh et al. (2024) The bacterial lysate OM-85 engages Toll-like receptors 2 and 4 triggering an immunomodulatory gene signature in human myeloid cells. Mucosal Immunology, 17:346–358. (Affiliations: IBR Bellinzona, OM Pharma, and Oncolines)
  • Nishiguchi et al. (2024) Selective CK1α degraders exert antiproliferative activity against a broad range of human cancer cell lines. Nature Communications, 15:482. (Affiliation: St. Jude Children’s Hospital)
  • Perera et al. (2023) Preclinical and emerging Phase 1 study data indicates that novel deuterated MET kinase inhibitor DO-2 mitigates the side effects seen with current approved MET kinase inhibitors. Poster presentation at AACR-NCI-EORTC Symposium on Molecular Targets and Cancer Therapeutics. (Affiliation: DeuterOncology N.V.)
  • Gupta et al. (2023) High SLFN11 expression correlates with sensitivity to lurbinectedin in small cell lung cancer (SCLC) models. Poster presentation at AACR Annual Meeting. (Affiliation: Jazz Pharmaceuticals, Inc.)
  • Gorter et al. (2023) Preclinical evaluation of MCLA-129, a bispecific antibody targeting EGFR and c-MET on solid tumor cells, in comparison with amivantamab. Poster presentation at AARC Annual Meeting 2023. (Affiliation: Merus N.V.)
  • King et al. (2022) Screening of NXP900 and dasatinib across 121 cancer cell lines identifies differences in their antiproliferative activity profiles. Poster presentation at EORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics. (Affiliations: University of Edinburgh, Cancer Research U.K., and Nuvectis Pharma Inc.)
  • Hughes et al. (2022) Uncovering the molecular mechanisms which predict sensitivity to a novel SRC kinase inhibitor NXP900 to inform personalized healthcare strategies. Poster presentation at AACR Annual Meeting 2022. (Affiliations: Cancer Research U.K., University of Edinburgh, and Nuvectis Pharma Inc.)
  • Lane et al. (2022) BAL0891: a novel, small molecule, dual TTK/PLK1 mitotic checkpoint inhibitor (MCI) with potent single agent activity. Poster presentation at ESMO TAT conference 2022. (Affiliation: Basilea Pharmaceutica International Ltd.)
  • Cordo et al. (2022) Phosophoproteomic profiling of T cell acute lymphoblastic leukemia reveals targetable kinases and combination treatment strategies. Nature Communications, 13:1048. (Affiliation: Prinses Máxima Center for Pediatric Oncology)
  • van der Zwet et al. (2021) MAPK-ERK is a central pathway in T-cell acute lymphoblastic leukemia that drives steroid resistance. Leukemia, 35 (12):3394-3405. (Affiliation: Prinses Máxima Center for Pediatric Oncology)
  • Beauchamp et al. (2020) Targeting N-myristoylation for therapy of B-cell lymphomas. Nature Communications, 11:5348. (Affiliations: University of Alberta and Pacylex Pharmaceuticals)
  • Grünewald et al. (2019) Rogaratinib: A potent and selective pan‐FGFR inhibitor with broad antitumor activity in FGFR‐overexpressing preclinical cancer models. International Journal of Cancer, 145 (5): 346–1357. (Affiliation: Bayer AG)
  • Gentile et al. (2018) A Novel interaction between the TLR7 and a colchicine derivative revealed through a computational and experimental Study. Pharmaceuticals, 11(1):22. (Affiliation: University of Alberta)
  • Wentsch et al. (2017) Optimized target residence time: type 1½ inhibitors for p38a MAP kinase with improved binding kinetics through direct interaction with the R-spine. Angewandte Chemie International Edition, 56(19):5363–5367. (Affiliations: University of Tübingen and NTRC)
  • Bohnacker et al. (2017) Deconvolution of Buparlisib’s mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic intervention. Nature Communications, 8:14683. (Affiliation: University of Basel)
  • Politz et al. (2017) Preclinical activity of the FGFR inhibitor rogaratinib (BAY 1163877) alone or in combination with antihormonal therapy in breast cancer. Cancer Research, 77(13 Supplement):1079. (Affiliation: Bayer AG)
  • Li et al. (2016) IL-7 receptor mutations and steroid resistance in pediatric T cell acute lymphoblastic leukemia: a genome sequencing study. PLoS Med, 13(12):e1002200. (Affiliations: Erasmus MC and NTRC)