Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Archives of Toxicology, Springer Science and Business Media LLC
    Type of Medium: Online Resource
    ISSN: 0340-5761 , 1432-0738
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2014
    detail.hit.zdb_id: 1458459-1
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Molecular Cancer, Springer Science and Business Media LLC, Vol. 14, No. 1 ( 2015-12)
    Type of Medium: Online Resource
    ISSN: 1476-4598
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2091373-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Global Food Security, Elsevier BV, Vol. 34 ( 2022-09), p. 100650-
    Type of Medium: Online Resource
    ISSN: 2211-9124
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 2682428-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Antimicrobial Resistance & Infection Control, Springer Science and Business Media LLC, Vol. 5, No. 1 ( 2016-12)
    Type of Medium: Online Resource
    ISSN: 2047-2994
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2016
    detail.hit.zdb_id: 2666706-X
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Molecular Cancer Therapeutics Vol. 18, No. 12_Supplement ( 2019-12-01), p. A065-A065
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12_Supplement ( 2019-12-01), p. A065-A065
    Abstract: Background: Advances in the culture of cells from human tissues have enabled the generation of normal and tumor organoid pairs derived from the same patient. These organoid cultures increase the scope for predicting responses to novel cancer therapeutics in patients. Here we present a high throughput 3D human intestinal organoid culture platform combined with high content image-based analysis. This platform allows visualization and quantification of responses beyond conventional cell viability measurements, including those associated with tumor killing, cell cycle arrest, toxicity and epithelial permeability and discriminates therapeutic and adverse responses in healthy tissue and tumor. Methods: Intestinal organoids, obtained from HUB Organoid Technology, cultured in natural extracellular matrix scaffolds show gene expression patterns, differentiation and functional characteristics that closely resemble the in vivo biology. To investigate the effect of standard-of-care (SoC) treatments, novel targeting antibodies and small molecules on tumor growth suppression and killing as well as function, formation and integrity of the gut epithelium, we tested a panel of normal and colorectal cancer organoids with a broad heterogeneity of mutations. 3D imaging and analysis was performed and responses in normal vs. tumor were compared. Results: High content 3D image analysis of the organoid enabled sensitive detection of treatment-induced and compound-specific morphological changes such as (inhibition of) growth, development, lumen formation, epithelial integrity and cell death. This enabled distinction between mechanisms of action and distinct effects and sensitivities in normal epithelium and tumor tissue from the same patient. Conclusion: 3D image analysis of in vitro cultured organoids represents a rapid and biologically relevant approach to test various anti-cancer-therapeutic modalities, including antibodies, antibody-drug conjugates and small molecules. Evaluating effects on healthy and tumor tissue from the same patient can give insight into potential toxicities and therapeutic window. Citation Format: Mariusz Madej, Cinthya Del Angel Zuvirie, Jara García Mateos, Leo Price, Lidia Daszkiewicz, Kuan Yan, Bram Herpers. Differential sensitivity of normal and tumor organoids to targeted therapies [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr A065. doi:10.1158/1535-7163.TARG-19-A065
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Developmental Cell, Elsevier BV, Vol. 12, No. 6 ( 2007-06), p. 901-915
    Type of Medium: Online Resource
    ISSN: 1534-5807
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2007
    detail.hit.zdb_id: 2053870-4
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 32-32
    Abstract: Background. Colorectal cancer (CRC) is the third most common cancer and remains a large unmet need. Dysregulation of Wnt and receptor tyrosine kinase (RTK) signalling pathways are important oncogenic driving events in CRC. Due to this dysregulation, Wnt target genes are expressed at higher levels in CRC particularly in tumor initiating cells. We previously performed an unbiased screen of bispecific antibodies (bAbs) targeting Wnt and RTK targets that resulted in the selection of MCLA-158. Methods. A cohort of 32 genetically and transcriptionally annotated patient-derived colorectal cancer and normal colon organoids were used to functionally characterize responses to antibodies based on morphological changes with high content 3D imaging. Binding affinity was measured by surface plasma resonance and cell based assays. The antibody binding epitopes were mapped by shotgun mutagenesis and FACS based screening. Ligand (R-spondin or EGF) blocking activity was measured in vitro by competition for ligand binding or functional inhibition of ligand dependent growth. In vivo activity was evaluated in xenograft models generated from organoids subcutaneously implanted into immunocompromised mice. Safety was evaluated via once weekly intravenous administration of MCLA-158 to cynomolgus monkeys for 4 weeks and monitoring for pathological changes. Results. MCLA-158, an ADCC enhanced common light chain IgG1 bispecific antibody, binds in domain III of EGFR and in the N-Cap/1st LRR of LGR5, both ligand binding regions, however, only EGF binding was blocked by MCLA-158. MCLA-158 demonstrated inhibitory activity in 74% of tumor organoids independent of KRAS mutational status but was not active on organoids of the cohort harboring both KRAS and PIK3CA mutations. MCLA-158 was significantly more active on organoids derived from tumors than from normal tissue in contrast to cetuximab, which demonstrated equivalent activity on both (range 20-100 fold, n=4). In vivo activity was evaluated against tumor organoids with different KRAS mutation status shown to be sensitive to MCLA-158 in vitro. In all cases, MCLA-158 significantly inhibited the growth of the tumor compared to both control and cetuximab treatment. Inhibitors of both the Wnt and EGFR pathways have shown significant toxicity in humans. An initial evaluation of MCLA-158 toxicity in cynomolgus monkeys did not demonstrate any pathological finding after repeated dosing at 25mg/kg. Conclusions. MCLA-158 demonstrates superior activity compared to reference antibodies in both in vitro and in vivo tumor organoid based assays regardless of KRAS status and was well tolerated in non-human primates. These preclinical data suggest MCLA-158 could benefit patients with metastatic CRC and warrant clinical evaluation. Citation Format: Rob Roovers, Bram Herpers, Mark James, Berina Eppink, Carme Cortina, David Maussang-Detaille, Ingrid Kolfschoten, Sylvia Boy, Marc van de Wetering, Wim De Lau, Robert Doornbos, Kuan Yan, Lucia Salinaro, Lex Bakker, john de Kruif, Hans Clevers, Robert Vries, Eduard Batlle, Leo Price, Mark Throsby. Preclinical evaluation of MCLA-158: A bispecific antibody targeting LGR5 and EGFR using patient-derived colon carcinoma organoids [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 32. doi:10.1158/1538-7445.AM2017-32
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 6315-6315
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 6315-6315
    Abstract: Background: The choice in advanced pre-clinical in vitro models that use cultures of patient-derived tissues has increased but their full potential in drug discovery has not been fully exploited. Assays often lack sensitivity or clinically relevant end-points or are not suitable for high throughput screening. Here we describe a 3D screening platform that combines complex biology with high content phenotype-based image analysis to provide visualization and quantification of the effects of various targeted therapies on healthy and diseased tissue. Improved characterization of compound effects increases the scope for predicting treatment responses in patients, discriminating different drug responses and flagging off-target effects. Methods: A panel of tumor and healthy intestinal organoids, obtained from HUB Organoid Technology, were cultured in natural extracellular matrix scaffolds that support appropriate gene expression, differentiation and functional characteristics of the tissue of origin. To investigate the effects of anti-EGFR targeted therapies in combination with standard-of-care (SoC) treatments on the function, formation and integrity of colorectal cancer organoids (CRCs) and matching normal gut epithelium, we set up the following intestinal organoid test systems: 1) A panel of CRCs with a broad heterogeneity of mutations for high throughput phenotype-based screening that enable accurate compound profiling 2) Normal vs. tumor organoid pair assays for off-target effect studies and 3) Mode of action studies in various tumoroid models. Results: High content 3D image analysis of the organoids enabled sensitive detection of treatment-induced and compound-specific morphological changes beyond conventional cell viability measurements, including those associated with tumor killing, cell cycle arrest, toxicity and epithelial integrity to discriminate therapeutic and adverse responses in intestinal organoids. This enabled distinction between different mechanisms of action and determination of compound synergy. The image-based measurements can be complemented with detection of secreted factors (e.g. cytokines, chemokines) or expressed genes in response to various therapeutic compounds. Conclusion: The advanced 3D image analysis of in vitro cultured organoids described here represents a rapid and biologically relevant approach to test various anti-cancer-therapeutic modalities, including antibodies, antibody-drug conjugates and small molecules. This platform technology is suitable not only for high throughput screening in organoid panels but also in-depth mode of action studies. Moreover, evaluating effects on healthy and tumor intestinal organoids from the same patient and between patients, gives insight into potential toxicities, therapeutic window and patient-derived differences in subsequent in vivo studies. Citation Format: Mariusz Madej, Cinthya Del Angel Zuvirie, Jara García Mateos, Leo Price, Kuan Yan, Bram Herpers. High content screening of intestinal organoid cultures to visualize and quantify effects of drug combinations [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6315.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. LB-044-LB-044
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. LB-044-LB-044
    Abstract: Background. There is very high attrition of compounds in early stage anti-cancer drug discovery. This is partly due to the failure of early stage drug testing platforms to represent human biology. 2D cultures of human cells lack the realistic complexity of an organism, leading to overestimation of compound potency. Animal models do retain complexity, but are limited in throughput and frequently fail to reflect human tumor biology. Patient-derived xenograft (PDX) models in immune-compromised mice allow propagation of and compound testing in human-derived tumors. To expand the potential of these human-relevant PDX models, we sought to develop 3D in vitro culture methods for PDX-derived tumor cells that show in vivo-like growth characteristics, invasion and responses to therapeutics. In combination with advanced 3D image analysis methods, we created a unique high throughput in vitro PDX screening platform that not only allows efficient identification of active and selective molecules but also enables selection of the optimal PDX tumor models for subsequent validation of candidates in vivo. Results. Each PDX model has its own unique growth characteristics. Hydrogel and growth media composition were optimized to support growth of tumor tissues in vitro from cells derived from bladder, gastric, breast, colon and lung cancer PDX tumors. Tumor tissues were cultured in a 384-well format and used to screen chemotherapeutics, small molecules, antibodies and antibody-drug-conjugates (ADCs). Using OcellO's 3D image analysis platform, Ominer, tumoroid growth, cell proliferation, apoptosis, invasion, cell polarity, differentiation and other aspects of cell and tissue architecture were analyzed and the effects of compound exposure on tumoroids was determined. By performing feature training based on reference compounds, we selected ±10 morphological features (out of more than 500) to generate a phenotypic signature that described the unique phenotypic change induced by each compound. Different compounds that target the same molecule were found to induce a similar morphological change whereas compounds with off-target effects could be discriminated. This approach enabled a high resolution evaluation and comparison of compound activity in an automated manner. Conclusions. We established several bladder, gastric, breast, colon and lung cancer PDX model-derived 3D tumor cultures in which standard-of-care and novel therapeutic agents (small molecules, antibodies and ADCs) can efficiently be screened, based on therapeutically relevant parameters and their changing morphological profile. This method enables both the in vitro selection of promising compounds in a pre-clinically relevant setting and the selection of optimum PDX tumor models for follow-up in vivo studies. This highly translational in vitro-in vivo PDX pipeline is expected to reduce attrition and increase efficiency in early drug-discovery. Citation Format: Bram Herpers, Lidia Daszkiewicz, Torsten Giesemann, Leo Price. A 384-well in vitro PDX culture platform for compound screening and PDX model selection. [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 LB-044.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4644-4644
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4644-4644
    Abstract: Ex vivo cultures of patient-derived tumor enable functional testing of treatment options and optimization of pre-clinical drug development. Patient biopsies or tumor from patient-derived xenografts (PDX) mouse models are a valuable source of human tumor material that can be cultured in 3D for screening of drug efficacy, drug resistance and cellular processes such as proliferation, survival and invasion. A high throughput approach using 384 well plates enables evaluation of multiple drug treatments and dose ranges in parallel. After compound exposure, cultures are fixed and stained with cellular markers. 3D image stack acquisition is followed by ultra-high content multiparametric analysis using the OMiner platform to profile drug responses and quantify tumor spheroid volume, apoptosis and tumor invasion. Dissected PDX material and tumor biopsy material, were used to establish 3D tumor cultures derived from various indications, including breast, ovarian, cervix, endometrium, stomach, pancreatic, colon, bladder and lung cancer. These were exposed to standard-of-care chemotherapeutics (e.g. 5-FU, taxanes, platinum compounds, anthracyclines, alkylating agents), small molecules (e.g. erlotinib, lapatinib, trametinib), targeted therapies (PARPi; niraparib, olaparib, rucaparib), antibodies (e.g. cetuximab, trastuzumab) and antibody-drug-conjugate (ADC, T-DM1) dose ranges. The tumor culture response was measured, generating dose-dependent profiles for relevant features. Patient-derived 3D tumor cultures were tested with standard-of-care and novel therapeutic agents and high content analysis was used to evaluate drug sensitivity. This method enables both the in vitro selection of drug candidates in a pre-clinical setting as well as efficient selection of PDX models for in vivo follow-up in the same tumor. This highly translational in vitro-in vivo PDX pipeline is expected to reduce attrition and increase efficiency in early drug-discovery. Correlation of drug sensitivity in 3D cultures from fresh patient tumor biopsies, on the other hand, can be used for development of predictive diagnostics and also provides a unique source of patient material for drug discovery and development. Citation Format: Thomas Dijkmans, Sander Basten, Bram Herpers, Kuan Yan, Torsten Giesemann, Julia Schueler, Willemijn Vader, Leo Price. Patient-derived 3D tumor cultures for clinical diagnostics and pre-clinical drug development [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4644.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages