17 results on '"Tijmen H. Booij"'
Search Results
2. Drug screening and genome editing in human pancreatic cancer organoids identifies drug-gene interactions and candidates for off-label therapy
- Author
-
Christian K. Hirt, Tijmen H. Booij, Linda Grob, Patrik Simmler, Nora C. Toussaint, David Keller, Doreen Taube, Vanessa Ludwig, Alexander Goryachkin, Chantal Pauli, Daniela Lenggenhager, Daniel J. Stekhoven, Christian U. Stirnimann, Katharina Endhardt, Femke Ringnalda, Lukas Villiger, Alexander Siebenhüner, Sofia Karkampouna, Marta De Menna, Janette Beshay, Hagen Klett, Marianna Kruithof-de Julio, Julia Schüler, and Gerald Schwank
- Subjects
pancreatic cancer ,organoids ,drug screening ,PDX ,BRCA2 ,ARID1A ,Genetics ,QH426-470 ,Internal medicine ,RC31-1245 - Abstract
Summary: Pancreatic cancer (PDAC) is a highly aggressive malignancy for which the identification of novel therapies is urgently needed. Here, we establish a human PDAC organoid biobank from 31 genetically distinct lines, covering a representative range of tumor subtypes, and demonstrate that these reflect the molecular and phenotypic heterogeneity of primary PDAC tissue. We use CRISPR-Cas9 genome editing and drug screening to characterize drug-gene interactions with ARID1A and BRCA2. We find that missense, but not frameshift, mutations in the PDAC driver gene ARID1A are associated with increased sensitivity to the kinase inhibitors dasatinib (p < 0.0001) and VE-821 (p < 0.0001). We further conduct an automated drug-repurposing screen with 1,172 FDA-approved compounds, identifying 26 compounds that effectively kill PDAC organoids, including 19 chemotherapy drugs currently approved for other cancer types. We validate the activity of these compounds in vitro and in vivo. The in vivo validated hits include emetine and ouabain, compounds that are approved for non-cancer indications and that perturb the ability of PDAC organoids to respond to hypoxia. Our study provides proof-of-concept for advancing precision oncology and for identifying candidates for drug repurposing via genome editing and drug screening in tumor organoid biobanks.
- Published
- 2022
- Full Text
- View/download PDF
3. Tumor Organoids as a Research Tool: How to Exploit Them
- Author
-
Tijmen H. Booij, Chiara M. Cattaneo, and Christian K. Hirt
- Subjects
tumor organoid culture ,personalized/precision medicine ,tumor microenvironment ,screening library ,high-throughput screening ,high-content screening ,Cytology ,QH573-671 - Abstract
Organoid models allow for the study of key pathophysiological processes such as cancer biology in vitro. They offer insights into all aspects covering tumor development, progression and response to the treatment of tissue obtained from individual patients. Tumor organoids are therefore not only a better tumor model than classical monolayer cell cultures but can be used as personalized avatars for translational studies. In this review, we discuss recent developments in using organoid models for cancer research and what kinds of advanced models, testing procedures and readouts can be considered.
- Published
- 2022
- Full Text
- View/download PDF
4. Patient-derived xenografts and organoids model therapy response in prostate cancer
- Author
-
Joel Grosjean, Peter Clark Gray, Jean-Philippe Theurillat, Sofia Karkampouna, Andrea Sboner, M. De Menna, M.R. De Filippo, George N. Thalmann, Andrea Garofoli, Eugenio Zoni, Andrej Benjak, Marco Bolis, Vera Genitsch, David Keller, Charlotte K.Y. Ng, Salvatore Piscuoglio, Irena Klima, Mark A. Rubin, Kenneth Eng, Arianna Vallerga, Tijmen H. Booij, Julio Marianna Kruithof-de, F. La Manna, Christian U. Stirnimann, and Mirjam Kiener
- Subjects
Oncology ,medicine.medical_specialty ,Prostate cancer ,Therapy response ,business.industry ,Urology ,Internal medicine ,Organoid ,medicine ,medicine.disease ,business - Published
- 2021
5. PD59-07 PERSONALISED ORGANOID DRUG TREATMENT AND THERAPY RESISTANCE ON NOVEL EARLY ONSET METASTASIS XENOGRAFT MODEL
- Author
-
Charlotte K.Y. Ng, Marta De Menna, Andrea Garofoli, Maria R. De Filippo, Salvatore Piscuoglio, Federico La Manna, Eugenio Zoni, Marianna Kruithof-de Julio, Andrea Sboner, Christian U. Stirnimann, Irena Klima, Joel Grosjean, Mark A. Rubin, Martin Spahn, Vera Genitsch, Tijmen H. Booij, Sofia Karkampouna, David Keller, and George N. Thalmann
- Subjects
Oncology ,medicine.medical_specialty ,Drug treatment ,business.industry ,Urology ,Internal medicine ,medicine ,Organoid ,Treatment resistance ,business ,medicine.disease ,Early onset ,Metastasis - Published
- 2020
6. Patient-derived xenografts and organoids model therapy response in prostate cancer
- Author
-
Mirjam Kiener, Marianna Kruithof-de Julio, George N. Thalmann, Salvatore Piscuoglio, Marta De Menna, Maria R. De Filippo, Federico La Manna, Peter C. Gray, Andrea Sboner, Eugenio Zoni, Charlotte K.Y. Ng, Jo eumll Grosjean, David Keller, Christian U. Stirnimann, Sofia Karkampouna, Marco Bolis, Tijmen H. Booij, Martin Spahn, Irena Klima, Mark A. Rubin, Kenneth Eng, Andrea Garofoli, Jean-Philippe Theurillat, and Vera Genitsch
- Subjects
Sorafenib ,Sunitinib ,Ponatinib ,Microsatellite instability ,SPOP ,Biology ,medicine.disease ,Primary tumor ,chemistry.chemical_compound ,Prostate cancer ,chemistry ,medicine ,Organoid ,Cancer research ,medicine.drug - Abstract
Therapy resistance and metastatic processes in prostate cancer (PCa) remain undefined, due to lack of experimental models that mimic different disease stages. We describe a novel androgen-dependent PCa patient-derived xenograft (PDX) model from treatment-naïve, soft tissue metastasis (PNPCa). RNA and whole-exome sequencing of the PDX tissue and organoids confirmed transcriptomic and genomic similarity to primary tumor. PNPCa harboursBRCA2 and CHD1somatic mutations, shows anSPOP/FOXA1-like transcriptomic signature and microsatellite instability, which occurs in 3% of advanced PCa and has never been modelledin vivo. Comparison of the treatment-naïve PNPCa with additional metastatic PDXs (BM18, LAPC9), in a medium-throughput organoid screen of FDA-approved compounds, revealed differential drug sensitivities. Multikinase inhibitors (ponatinib, sunitinib, sorafenib) were broadly effective on all PDX- and patient-derived organoids from advanced cases with acquired resistance to standard-of-care compounds. This proof-of-principle study may provide a preclinical tool to screen drug responses to standard-of-care and newly identified, repurposed compounds.
- Published
- 2020
7. Patient-derived xenografts and organoids model therapy response in prostate cancer
- Author
-
Marco Bolis, Arianna Vallerga, Salvatore Piscuoglio, Andrej Benjak, Andrea Sboner, Federico La Manna, Mirjam Kiener, Joel Grosjean, Tijmen H. Booij, David Keller, Charlotte K.Y. Ng, Eugenio Zoni, Martin Spahn, Vera Genitsch, Sofia Karkampouna, Irena Klima, Mark A. Rubin, Kenneth Eng, Marianna Kruithof-de Julio, Christian U. Stirnimann, Jean-Philippe Theurillat, Andrea Garofoli, Maria R. De Filippo, Marta De Menna, George N. Thalmann, and Peter C. Gray
- Subjects
0301 basic medicine ,Sorafenib ,Male ,Science ,Medizin ,General Physics and Astronomy ,610 Medicine & health ,Antineoplastic Agents ,SPOP ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cancer screening ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,medicine ,Organoid ,Humans ,Neoplasm Metastasis ,Cancer models ,Multidisciplinary ,Sunitinib ,business.industry ,Genome, Human ,Cancer stem cells ,Ponatinib ,Microsatellite instability ,Prostatic Neoplasms ,General Chemistry ,medicine.disease ,Primary tumor ,Xenograft Model Antitumor Assays ,3. Good health ,Organoids ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Androgens ,570 Life sciences ,biology ,business ,Transcriptome ,medicine.drug - Abstract
Therapy resistance and metastatic processes in prostate cancer (PCa) remain undefined, due to lack of experimental models that mimic different disease stages. We describe an androgen-dependent PCa patient-derived xenograft (PDX) model from treatment-naïve, soft tissue metastasis (PNPCa). RNA and whole-exome sequencing of the PDX tissue and organoids confirmed transcriptomic and genomic similarity to primary tumor. PNPCa harbors BRCA2 and CHD1 somatic mutations, shows an SPOP/FOXA1-like transcriptomic signature and microsatellite instability, which occurs in 3% of advanced PCa and has never been modeled in vivo. Comparison of the treatment-naïve PNPCa with additional metastatic PDXs (BM18, LAPC9), in a medium-throughput organoid screen of FDA-approved compounds, revealed differential drug sensitivities. Multikinase inhibitors (ponatinib, sunitinib, sorafenib) were broadly effective on all PDX- and patient-derived organoids from advanced cases with acquired resistance to standard-of-care compounds. This proof-of-principle study may provide a preclinical tool to screen drug responses to standard-of-care and newly identified, repurposed compounds., To date, patients still succumb to cancer, due to tumors not responding to therapy or ultimately acquiring resistance. Here the authors show that by exploiting patient derived organoids and a treatment-naïve patient derived xenograft, patient therapy can be personalized.
- Published
- 2020
8. Personalised organoid drug treatment and therapy resistance characterization based on novel BRCA2 prostate cancer xenograft of SPOP-like phenotype and microsatellite instability
- Author
-
Joel Grosjean, M.R. De Filippo, M. De Menna, Salvatore Piscuoglio, F. La Manna, G.N. Thalmann, M. Kruithof-De Julio, Tijmen H. Booij, Christian U. Stirnimann, Sofia Karkampouna, Eugenio Zoni, Charlotte K.Y. Ng, David Keller, Irena Klima, Andrea Sboner, Andrea Garofoli, Martin Spahn, Vera Genitsch, and A.R. Mark
- Subjects
business.industry ,Urology ,Microsatellite instability ,SPOP ,lcsh:Diseases of the genitourinary system. Urology ,lcsh:RC870-923 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Phenotype ,Drug treatment ,Prostate cancer ,Cancer research ,Organoid ,Medicine ,Treatment resistance ,business - Published
- 2020
9. In vitro 3D phenotypic drug screen identifies celastrol as an effective in vivo inhibitor of polycystic kidney disease
- Author
-
Wouter N. Leonhard, Leo S. Price, Tijmen H. Booij, Dorien J.M. Peters, Hester Bange, Kimberley Veraar, Johannes G. Dauwerse, Kuan Yan, Gerard J. P. van Westen, Anna J. Plugge, Michiel Fokkelman, and Bob van de Water
- Subjects
0301 basic medicine ,Phenotypic screening ,Drug Evaluation, Preclinical ,Pharmacology ,Biology ,Kidney Function Tests ,AcademicSubjects/SCI01180 ,Kidney cysts ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Pyrvinium pamoate ,0302 clinical medicine ,In vivo ,Genetics ,medicine ,Polycystic kidney disease ,Animals ,Cyst ,Molecular Biology ,Polycystic Kidney Diseases ,Kidney ,Cysts ,PKD ,High-content screening ,Articles ,Cell Biology ,General Medicine ,Celastrol ,medicine.disease ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,medicine.symptom ,Pentacyclic Triterpenes ,Signal Transduction ,3D - Abstract
Polycystic kidney disease (PKD) is a prevalent genetic disorder, characterized by the formation of kidney cysts that progressively lead to kidney failure. The currently available drug tolvaptan is not well tolerated by all patients and there remains a strong need for alternative treatments. The signaling rewiring in PKD that drives cyst formation is highly complex and not fully understood. As a consequence, the effects of drugs are sometimes difficult to predict. We previously established a high throughput microscopy phenotypic screening method for quantitative assessment of renal cyst growth. Here, we applied this 3D cyst growth phenotypic assay and screened 2320 small drug-like molecules, including approved drugs. We identified 81 active molecules that inhibit cyst growth. Multi-parametric phenotypic profiling of the effects on 3D cultured cysts discriminated molecules that showed preferred pharmacological effects above genuine toxicological properties. Celastrol, a triterpenoid from Tripterygium Wilfordii, was identified as a potent inhibitor of cyst growth in vitro. In an in vivo iKspCre-Pkd1lox,lox mouse model for PKD, celastrol inhibited the growth of renal cysts and maintained kidney function.
- Published
- 2019
10. 3D Cell-Based Assays for Drug Screens: Challenges in Imaging, Image Analysis, and High-Content Analysis
- Author
-
Erik H.J. Danen, Leo S. Price, and Tijmen H. Booij
- Subjects
0301 basic medicine ,Quality Control ,3D culture ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Drug Evaluation, Preclinical ,Antineoplastic Agents ,Computational biology ,Review ,high-content screening ,Biochemistry ,Analytical Chemistry ,03 medical and health sciences ,3D cell culture ,0302 clinical medicine ,In vivo ,image analysis ,Cell Line, Tumor ,Drug Discovery ,Image Processing, Computer-Assisted ,Humans ,Automation, Laboratory ,Microscopy, Confocal ,Drug discovery ,Drug screens ,imaging ,Cell based assays ,Pipeline (software) ,3. Good health ,High-Throughput Screening Assays ,030104 developmental biology ,Drug development ,030220 oncology & carcinogenesis ,High-content screening ,Molecular Medicine ,Biotechnology - Abstract
The introduction of more relevant cell models in early preclinical drug discovery, combined with high-content imaging and automated analysis, is expected to increase the quality of compounds progressing to preclinical stages in the drug development pipeline. In this review we discuss the current switch to more relevant 3D cell culture models and associated challenges for high-throughput screening and high-content analysis. We propose that overcoming these challenges will enable front-loading the drug discovery pipeline with better biology, extracting the most from that biology, and, in general, improving translation between in vitro and in vivo models. This is expected to reduce the proportion of compounds that fail in vivo testing due to a lack of efficacy or to toxicity.
- Published
- 2019
11. Aven-mediated checkpoint kinase control regulates proliferation and resistance to chemotherapy in conventional osteosarcoma
- Author
-
Tijmen H Booij, Anne-Marie Cleton-Jansen, Zuzanna Baranski, Pancras C.W. Hogendoorn, Erik H.J. Danen, Bob van de Water, Judith V.M.G. Bovée, and Leo S. Price
- Subjects
musculoskeletal diseases ,Chemotherapy ,animal structures ,Kinase ,medicine.medical_treatment ,Biology ,medicine.disease ,environment and public health ,Conventional Osteosarcoma ,Pathology and Forensic Medicine ,enzymes and coenzymes (carbohydrates) ,medicine.anatomical_structure ,Immunology ,medicine ,Cancer research ,Osteosarcoma ,Gene silencing ,Doxorubicin ,Sarcoma ,biological phenomena, cell phenomena, and immunity ,Sensitization ,medicine.drug - Abstract
Conventional high-grade osteosarcoma is the most common primary bone sarcoma, with relatively high incidence in young people. In this study we found that expression of Aven correlates inversely with metastasis-free survival in osteosarcoma patients and is increased in metastases compared to primary tumours. Aven is an adaptor protein that has been implicated in anti-apoptotic signalling and serves as an oncoprotein in acute lymphoblastic leukaemia. In osteosarcoma cells, silencing Aven triggered G2 cell-cycle arrest; Chk1 protein levels were attenuated and ATR-Chk1 DNA damage response signalling in response to chemotherapy was abolished in Aven-depleted osteosarcoma cells, while ATM, Chk2 and p53 activation remained intact. Osteosarcoma is notoriously difficult to treat with standard chemotherapy, and we examined whether pharmacological inhibition of the Aven-controlled ATR-Chk1 response could sensitize osteosarcoma cells to genotoxic compounds. Indeed, pharmacological inhibitors targeting Chk1/Chk2 or those selective for Chk1 synergized with standard chemotherapy in 2D cultures. Likewise, in 3D extracellular matrix-embedded cultures, Chk1 inhibition led to effective sensitization to chemotherapy. Together, these findings implicate Aven in ATR-Chk1 signalling and point towards Chk1 inhibition as a strategy to sensitize human osteosarcomas to chemotherapy.
- Published
- 2015
12. MEK inhibition induces apoptosis in osteosarcoma cells with constitutive ERK1/2 phosphorylation
- Author
-
Yvonne de Jong, Erik H.J. Danen, Leo S. Price, Zuzanna Baranski, Tijmen H Booij, Marieke L. Kuijjer, Anne-Marie Cleton-Jansen, Pancras C.W. Hogendoorn, Judith V.M.G. Bovée, and Bob van de Water
- Subjects
MAPK/ERK pathway ,Trametinib ,3D culture ,Cancer Research ,medicine.diagnostic_test ,Biology ,medicine.disease ,Molecular biology ,MEK ,Western blot ,Cell culture ,Apoptosis ,osteosarcoma ,pharmacological inhibition ,Genetics ,medicine ,Osteosarcoma ,Viability assay ,ERK phosphorylation ,Protein kinase B ,Research Paper - Abstract
Conventional high-grade osteosarcoma is the most common primary bone cancer with relatively high incidence in young people. Recurrent and metastatic tumors are difficult to treat. We performed a kinase inhibitor screen in two osteosarcoma cell lines, which identified MEK1/2 inhibitors. These inhibitors were further validated in a panel of six osteosarcoma cell lines. Western blot analysis was performed to assess ERK activity and efficacy of MEK inhibition. A 3D culture system was used to validate results from 2D monolayer cultures. Gene expression analysis was performed to identify differentially expressed gene signatures in sensitive and resistant cell lines. Activation of the AKT signaling network was explored using Western blot and pharmacological inhibition. In the screen, Trametinib, AZD8330 and TAK-733 decreased cell viability by more than 50%. Validation in six osteosarcoma cell lines identified three cell lines as resistant and three as sensitive to the inhibitors. Western blot analysis of ERK activity revealed that sensitive lines had high constitutive ERK activity. Treatment with the three MEK inhibitors in a 3D culture system validated efficacy in inhibition of osteosarcoma viability. MEK1/2 inhibition represents a candidate treatment strategy for osteosarcomas displaying high MEK activity as determined by ERK phosphorylation status.
- Published
- 2015
13. High-Throughput Phenotypic Screening of Kinase Inhibitors to Identify Drug Targets for Polycystic Kidney Disease
- Author
-
Johannes G. Dauwerse, Yu Qin, Steven J. Kunnen, Gerard J. P. van Westen, Dorien J.M. Peters, Michiel Fokkelman, Kuan Yan, Tijmen H Booij, Hester Bange, Bob van de Water, Wouter N. Leonhard, and Leo S. Price
- Subjects
0301 basic medicine ,3D culture ,Phenotypic screening ,030232 urology & nephrology ,Pharmacology ,Biochemistry ,Hydrogel, Polyethylene Glycol Dimethacrylate ,Analytical Chemistry ,Cell Line ,03 medical and health sciences ,Mice ,Phosphatidylinositol 3-Kinases ,high-content screening (HCS) ,0302 clinical medicine ,Aurora kinase ,Cyclin-dependent kinase ,high-throughput screening (HTS) ,Polycystic kidney disease ,medicine ,Animals ,kinase inhibitors ,Cyst ,Molecular Targeted Therapy ,Kidney Tubules, Collecting ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Original Research ,Phosphoinositide-3 Kinase Inhibitors ,Polycystic Kidney Diseases ,polycystic kidney disease ,biology ,Drug discovery ,TOR Serine-Threonine Kinases ,Colforsin ,In vitro toxicology ,medicine.disease ,3. Good health ,High-Throughput Screening Assays ,030104 developmental biology ,Phenotype ,biology.protein ,Cancer research ,Molecular Medicine ,Biotechnology - Abstract
Polycystic kidney disease (PKD) is a prevalent disorder characterized by renal cysts that lead to kidney failure. Various signaling pathways have been targeted to stop disease progression, but most interventions still focus on alleviating PKD-associated symptoms. The mechanistic complexity of the disease, as well as the lack of functional in vitro assays for compound testing, has made drug discovery for PKD challenging. To identify modulators of PKD, Pkd1–/– kidney tubule epithelial cells were applied to a scalable and automated 3D cyst culture model for compound screening, followed by phenotypic profiling to determine compound efficacy. We used this screening platform to screen a library of 273 kinase inhibitors to probe various signaling pathways involved in cyst growth. We show that inhibition of several targets, including aurora kinase, CDK, Chk, IGF-1R, Syk, and mTOR, but, surprisingly, not PI3K, prevented forskolin-induced cyst swelling. Additionally, we show that multiparametric phenotypic classification discriminated potentially undesirable (i.e., cytotoxic) compounds from molecules inducing the desired phenotypic change, greatly facilitating hit selection and validation. Our findings show that a pathophysiologically relevant 3D cyst culture model of PKD coupled to phenotypic profiling can be used to identify potentially therapeutic compounds and predict and validate molecular targets for PKD.
- Published
- 2017
14. Development of a 3D Tissue Culture-Based High-Content Screening Platform That Uses Phenotypic Profiling to Discriminate Selective Inhibitors of Receptor Tyrosine Kinases
- Author
-
Kuan Yan, György Kéri, Laszlo Orfi, Bálint Szokol, Leo S. Price, Tijmen H Booij, Maarten Klop, Csaba Szántai-Kis, and Bob van de Water
- Subjects
0301 basic medicine ,Male ,C-Met ,Special Collection ,EGFR ,phenotypic profiling ,Biology ,high-content screening ,Biochemistry ,Receptor tyrosine kinase ,Analytical Chemistry ,Small Molecule Libraries ,03 medical and health sciences ,3D cell culture ,chemistry.chemical_compound ,Epidermal growth factor ,Humans ,Protein Kinase Inhibitors ,3D tissue culture ,Cell Proliferation ,c-Met ,Prostatic Neoplasms ,Hydrogels ,Proto-Oncogene Proteins c-met ,Molecular biology ,High-Throughput Screening Assays ,ErbB Receptors ,030104 developmental biology ,chemistry ,Cell culture ,High-content screening ,Cancer cell ,Cancer research ,biology.protein ,Molecular Medicine ,Drug Screening Assays, Antitumor ,Tyrosine kinase ,Biotechnology - Abstract
3D tissue cultures provide a more physiologically relevant context for the screening of compounds, compared with 2D cell cultures. Cells cultured in 3D hydrogels also show complex phenotypes, increasing the scope for phenotypic profiling. Here we describe a high-content screening platform that uses invasive human prostate cancer cells cultured in 3D in standard 384-well assay plates to study the activity of potential therapeutic small molecules and antibody biologics. Image analysis tools were developed to process 3D image data to measure over 800 phenotypic parameters. Multiparametric analysis was used to evaluate the effect of compounds on tissue morphology. We applied this screening platform to measure the activity and selectivity of inhibitors of the c-Met and epidermal growth factor (EGF) receptor (EGFR) tyrosine kinases in 3D cultured prostate carcinoma cells. c-Met and EGFR activity was quantified based on the phenotypic profiles induced by their respective ligands, hepatocyte growth factor and EGF. The screening method was applied to a novel collection of 80 putative inhibitors of c-Met and EGFR. Compounds were identified that induced phenotypic profiles indicative of selective inhibition of c-Met, EGFR, or bispecific inhibition of both targets. In conclusion, we describe a fully scalable high-content screening platform that uses phenotypic profiling to discriminate selective and nonselective (off-target) inhibitors in a physiologically relevant 3D cell culture setting.
- Published
- 2016
15. Aven-mediated checkpoint kinase control regulates proliferation and resistance to chemotherapy in conventional osteosarcoma
- Author
-
Zuzanna, Baranski, Tijmen H, Booij, Anne-Marie, Cleton-Jansen, Leo S, Price, Bob, van de Water, Judith V M G, Bovée, Pancras C W, Hogendoorn, and Erik H J, Danen
- Subjects
Osteosarcoma ,Antibiotics, Antineoplastic ,Gene Expression Profiling ,Membrane Proteins ,Apoptosis ,Bone Neoplasms ,Ataxia Telangiectasia Mutated Proteins ,Thiophenes ,G2 Phase Cell Cycle Checkpoints ,Gene Expression Regulation, Neoplastic ,Doxorubicin ,Cell Line, Tumor ,Checkpoint Kinase 1 ,Humans ,Urea ,RNA Interference ,Phosphorylation ,Apoptosis Regulatory Proteins ,Protein Kinases ,Adaptor Proteins, Signal Transducing ,DNA Damage ,Oligonucleotide Array Sequence Analysis ,Signal Transduction - Abstract
Conventional high-grade osteosarcoma is the most common primary bone sarcoma, with relatively high incidence in young people. In this study we found that expression of Aven correlates inversely with metastasis-free survival in osteosarcoma patients and is increased in metastases compared to primary tumours. Aven is an adaptor protein that has been implicated in anti-apoptotic signalling and serves as an oncoprotein in acute lymphoblastic leukaemia. In osteosarcoma cells, silencing Aven triggered G2 cell-cycle arrest; Chk1 protein levels were attenuated and ATR-Chk1 DNA damage response signalling in response to chemotherapy was abolished in Aven-depleted osteosarcoma cells, while ATM, Chk2 and p53 activation remained intact. Osteosarcoma is notoriously difficult to treat with standard chemotherapy, and we examined whether pharmacological inhibition of the Aven-controlled ATR-Chk1 response could sensitize osteosarcoma cells to genotoxic compounds. Indeed, pharmacological inhibitors targeting Chk1/Chk2 or those selective for Chk1 synergized with standard chemotherapy in 2D cultures. Likewise, in 3D extracellular matrix-embedded cultures, Chk1 inhibition led to effective sensitization to chemotherapy. Together, these findings implicate Aven in ATR-Chk1 signalling and point towards Chk1 inhibition as a strategy to sensitize human osteosarcomas to chemotherapy.
- Published
- 2014
16. Abstract 1703: A 3D tissue culture-based utra-high content screening platform that uses phenotypic profiling of cancer tissues to identify selective inhibitors of receptor tyrosine kinases
- Author
-
Tijmen H Booij, Maarten Klop, and Leo S. Price
- Subjects
Cancer Research ,biology ,Biological activity ,Molecular biology ,Receptor tyrosine kinase ,Tissue culture ,Oncology ,Cell culture ,High-content screening ,biology.protein ,Cancer research ,Receptor ,Tyrosine kinase ,EGFR inhibitors - Abstract
3D tissue cultures provide a more physiologically relevant context for the screening of compounds, compared to 2D cell cultures. Cells cultured in 3D hydrogels also show complex phenotypes, increasing the scope for phenotypic profiling. Here we describe a high-content screening platform that uses human micro-tissues cultured in 3D in standard 384 well assay plates to study the activity of biologically active molecules. Image analysis tools were developed to process 3D image data to measure 854 phenotype-derived parameters. Multiparametric analysis was used to evaluate the effect of compounds on tissue morphology. We applied this screening platform to measure the activity and selectivity of inhibitors of the c-Met and EGF receptor (EGFR) tyrosine kinases in 3D cultured prostate carcinoma cells. c-Met and EGFR activity was quantified based on the invasive phenotypic profiles induced by their respective ligands, HGF and EGF, which were added in parallel assays. c-Met and EGFR inhibitors generally showed the expected inhibitory profile equivalent to that obtained in the absence of cognate ligand. However, one phase-III c-Met inhibitor, displayed an unexpected phenotypic profile characterised by potent inhibition of tumour cell invasion and off-target effects consistent with a previously reported polypharmacology of this compound. The screening method was applied to a novel collection of 70 bi-specific compounds designed to target both c-Met and EGFR. Compounds were identified that induced phenotypic profiles indicative of selective inhibition of either c-Met, EGFR or both targets. In conclusion, this poster describes a fully scalable high-content screening platform that uses phenotypic profiling to evaluate receptor tyrosine kinase inhibitor selectivity and activity in a physiologically relevant setting. Citation Format: Leo S. Price, Tijmen Booij, Maarten Klop. A 3D tissue culture-based utra-high content screening platform that uses phenotypic profiling of cancer tissues to identify selective inhibitors of receptor tyrosine kinases. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1703. doi:10.1158/1538-7445.AM2015-1703
- Published
- 2015
17. Abstract 3780: Aven-mediated checkpoint kinase control regulates proliferation and resistance to chemotherapy in osteosarcoma cells
- Author
-
Zuzanna Baranski, Leo S. Price, Tijmen H Booij, Pancras C.W. Hogendoorn, Anne-Marie Cleton, Judith V.M.G. Bovée, Erik H.J. Danen, Jolieke G. van Oosterwijk, Yvonne de Jong, and Bob van de Water
- Subjects
Cancer Research ,Chemotherapy ,DNA damage ,Kinase ,business.industry ,medicine.medical_treatment ,Cancer ,Bone Sarcoma ,medicine.disease ,Oncology ,Immunology ,Cancer research ,medicine ,Gene silencing ,Osteosarcoma ,biological phenomena, cell phenomena, and immunity ,Chondrosarcoma ,business - Abstract
Osteosarcoma is the most common primary bone malignancy with relatively high incidence in young people. Here, we found that expression of Aven was inversely correlated with metastasis-free survival in osteosarcoma patients and is increased in metastases compared to primary osteosarcoma biopsies. Also in chondrosarcoma, the second most common bone sarcoma, expression correlated with increased histological grade. Aven is an adaptor protein that has been implicated in anti-apoptotic signaling and serves as an oncoprotein in acute lymphoblastic leukemia. In human osteosarcoma cells, silencing Aven triggered a G2 cell cycle arrest. Chk1 protein levels were attenuated and ATR-Chk1 DNA damage response signaling in response to chemotherapy was abolished in Aven-depleted osteosarcoma cells while ATM, Chk2, and p53 activation remained intact. Osteosarcoma is notoriously difficult to treat with standard chemotherapy, and we examined whether pharmacological inhibition of the Aven-controlled ATR-Chk1 response could sensitize osteosarcoma cells to genotoxic compounds. Indeed, pharmacological inhibitors targeting Chk1/Chk2 or those selective for Chk1 synergized with standard chemotherapy in 2D cultures. Likewise, in 3D extracellular matrix-embedded cultures Chk1 inhibition effectively sensitized human osteosarcoma cells to chemotherapy. Together, these findings implicate Aven in ATR-Chk1 signaling and point towards Chk1 inhibition as a strategy to sensitize osteosarcomas to chemotherapy. Citation Format: Zuzanna Baranski, Tijmen T.H. Booij, Yvonne de Jong, Jolieke van Oosterwijk, Anne-Marie Cleton, Leo Price, Bob van de Water, Judith V.M.G. Bovée, Pancras C.W. Hogendoorn, Erik Danen. Aven-mediated checkpoint kinase control regulates proliferation and resistance to chemotherapy in osteosarcoma cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3780. doi:10.1158/1538-7445.AM2015-3780
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.