Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 12 ( 2021-10-6)
    Abstract: Colorectal cancer (CRC) is the third most diagnosed malignancy and the second leading cause of cancer-related deaths worldwide. Locally advanced and metastatic disease exhibit resistance to therapy and are prone to recurrence. Despite significant advances in standard of care and targeted (immuno)therapies, the treatment effects in metastatic CRC patients have been modest. Untreatable cancer metastasis accounts for poor prognosis and most CRC deaths. The generation of a strong immunosuppressive tumor microenvironment (TME) by CRC constitutes a major hurdle for tumor clearance by the immune system. Dendritic cells (DCs), often impaired in the TME, play a critical role in the initiation and amplification of anti-tumor immune responses. Evidence suggests that tumor-mediated DC dysfunction is decisive for tumor growth and metastasis initiation, as well as for the success of immunotherapies. Unravelling and understanding the complex crosstalk between CRC and DCs holds promise for identifying key mechanisms involved in tumor progression and spread that can be exploited for therapy. The main goal of this review is to provide an overview of the current knowledge on the impact of CRC-driven immunosuppression on DCs phenotype and functionality, and its significance for disease progression, patient prognosis, and treatment response. Moreover, present knowledge gaps will be highlighted as promising opportunities to further understand and therapeutically target DC dysfunction in CRC. Given the complexity and heterogeneity of CRC, future research will benefit from the use of patient-derived material and the development of in vitro organoid-based co-culture systems to model and study DCs within the CRC TME.
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2021
    detail.hit.zdb_id: 2606827-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 987-987
    Abstract: Background: Distant metastatic disease remains the main cause of colorectal cancer (CRC) mortality, but our knowledge on how cancer cells travel from the primary tumor to a secondary site is limited. Lymph node metastases (LNM) play a central role in the staging of CRC patients since they are currently viewed as the gateway to distant metastases. However, in recent years additional forms of locoregional spread with varying prognostic impact have been identified, with tumor deposits (TD) having significantly more impact than LNM. The biological explanation for the difference in prognostic impact between LNM and TD is lacking, while this is essential for a more comprehensive understanding of the role of these biomarkers CRC spread. Therefore, in this study, shotgun proteomics were used to compare the biological phenotype of TD and LNM. Methods: From 12 CRC patients, one TD and one LNM were included for paired protein analyses. Proteins were isolated from formalin-fixed paraffin-embedded tissue after which a filter aided sample preparation method was performed. Then, the samples were analyzed by liquid-chromatography tandem mass-spectrometry, and after data handling and filtering, 5578 differentially expressed proteins were used for downstream analyses. Differences in expression were visualized using heatmaps and volcano plots, and enrichment analyses as well as gene set enrichment was performed. All analyses were performed using R version 4.1.2. Results: Among the main findings was that the proteins ASPN, MRC2, SPON1, MXRA5, BGN, LUM, and PLOD2 were overexpressed in TD. These proteins play a role in stimulating pro-tumorigenic processes such as migration, invasion, and epithelial to mesenchymal transition. For the enrichment analyses all proteins with a Fold change of less than -1 and more than 1 were included. Enrichment analyses using the KEGG pathways showed more immune activity in the LNM, with upregulated B-cell and T-cell signaling (p & lt;0.001), while TD showed more interaction with the extracellular matrix in the form of proteoglycans, focal adhesion, and ECM-receptor interaction (p & lt;0.01). In addition, when hallmarks from the Molecular Signatures Database (MSigDB) were used for the enrichment analyses, TD showed the hallmark of epithelial mesenchymal transition (p & lt;0.005). Conclusions: This study shows that TD have a more aggressive and invasive phenotype compared to LNM on protein level. TD had a higher expression of proteins that promote pro-tumorigenic processes and a more extensive interaction with the extracellular matrix. Furthermore, the hallmark of epithelial mesenchymal transition is enriched in TD compared to LNM. These findings form a possible explanation for the strong prognostic impact of TD, and give insight into the heterogeneity of different modes of locoregional spread in CRC. Citation Format: Nelleke P. Brouwer, Loth Webbink, Shannon Van Lent-Van Vliet, Pascal W. Jansen, Femke Simmer, Daniele V. Tauriello, Michiel Vermeulen, Iris D. Nagtegaal. Proteomics identifies differences in the biological phenotype of tumor deposits and lymph node metastases in colorectal cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 987.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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 ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 418-418
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 418-418
    Abstract: For unresectable metastatic colorectal cancer (mCRC), the main treatment strategy is combination chemotherapy, targeted therapy, or immunotherapy. Long-term treatment benefit of these therapies is limited to a small subgroup of patients. To improve treatment outcome, many tyrosine kinase inhibitors (TKIs) have been clinically tested in patients with mCRC. Most failed already in early-phase clinical trials due to intrinsic resistance. ATP Binding Cassette Transporter (ABC)B1- and ABCG2-transporters are known contributors to treatment resistance in mCRC. Therefore, we evaluated whether inhibition of these transporters could potentiate the efficacy of TKIs in colorectal cancer (CRC) models. CRC cell lines, HT29, COLO320 and CACO2 and newly established patient-derived tumor organoids (PDTOs) from mCRC biopsies were first analyzed for the presence of ABCB1 and ABCG2 expression. Models that expressed ABCB1 or ABCG2 were pre-incubated with a non-toxic dose of elacridar, a potent transporter inhibitor, and exposed to a concertation range of three TKIs; sunitinib, cediranib and osimertinib. Next, we measured intracellular drug concentrations using LC/MS-MS one hour after TKI exposure (20 µM) with or without pre-incubation of elacridar. Using the fluorescent properties of sunitinib, we performed live-cell imaging to compare intracellular sunitinib distribution in cells that were or were not pre-incubated with elacridar. ABCB1 and ABCG2 expression was observed in all CRC models except for HT29 cells, which did not express ABCB1. Pre-incubation with elacridar significantly improved sunitinib efficacy in CRC cells. The IC50 decreased from 2.15 µM to 1.17 µM (p = 0.04), from 1.32 µM to 0.44 µM (p = 0.03), and from 4.94 µM to 3.43 µM (p = 0.03) for HT29, COLO320 and CACO2 cells respectively. This effect was not observed for cediranib and osimertinib. Although not significant, for both PDTO models tested, the IC50 for sunitinib decreased from 8.04 µM to 3.29 µM and from 6.93 µM to 4.16 µM. Intracellular sunitinib concentrations in the tested CRC cell models after sunitinib exposure ranged from 2.46 mM to 4.13 mM without elacridar and from 0.38 mM µM to 1.31 mM when pre-incubated with elacridar (p & lt; 0.01). Live-cell imaging of CRC cells revealed a different pattern of intracellular sunitinib accumulation with and without pre-incubation with elacridar. Inhibition of multi-drug resistance proteins ABCB1 and ABCG2 with the clinically tested selective transporter inhibitor elacridar potentiates sunitinib efficacy in CRC models. Interestingly, this was accompanied with reduced intracellular sunitinib accumulation. Further research in mCRC PDTOs is warranted to confirm whether this mechanism is involved in intrinsic TKI resistance and therefore contribute to the early-phase failure of clinical trials testing TKIs for mCRC. Citation Format: Dennis Poel, Kirti K. Iyer, Bob van Gasteren, Beau Dagniaux, Erik van den Hombergh, Nielka P. van Erp, Daniele V. Tauriello, Henk M. Verheul. Elacridar potentiates sunitinib efficacy in colorectal cancer models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 418.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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 ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4028-4028
    Abstract: Despite major interest in tyrosine kinase inhibitors (TKIs) as a treatment option for metastatic colorectal cancer (mCRC), almost all TKIs tested for mCRC fail in early-phase clinical trials. Although showing specific target inhibition at low concentrations, TKIs have a much broader kinase inhibitory potency at higher concentrations. Alternative high dose regimens have been proposed to explore if efficacy can be improved with acceptable toxicity. We used 3D matrix-embedded tumor organoids as a preclinical platform to determine optimal drug exposure, i.e. preclinical pharmacology, and to dissect the mechanisms of action to potentially convert the dismal translational success rate of TKIs for mCRC. We established patient-derived tumor organoids (PDTOs) from mCRC biopsies and, based on favorable physicochemical and pharmacokinetic properties, selected 3 TKIs (sunitinib, cediranib and osimertinib). Following standard IC50 assessment using continuous dosing with a concentration range, we investigated the cytotoxic antitumor effect of high-dose, short-exposure (HDSE) treatment. Five PDTOs were exposed to 20 µM TKI for 1-24h, washed and given normal medium, and PDTO-outgrowth was determined 1 week later. At exposures of 1, 3 and 6h, we measured intra-tumoroid TKI concentrations using a clinically validated LC/MS-MS method. PDTO cell death was observed using live-cell microscopy, and quantified by both caspase 3/7 enzyme activity assay and cleaved caspase-3 immunofluorescent staining. While PDTOs could be categorized for their sensitivity across tested TKIs, all were highly sensitive for osimertinib (IC50 values 0.40-3.8 µM). Lower sensitivity was observed for sunitinib (2.0-10.5 µM) and cediranib (2.5-7.1 µM). Only for osimertinib exposure to 20 µM for 3h was sufficient to block proliferation in all PDTOs. Interestingly, peak intra-tumoroid TKI concentration measurements across PDTOs revealed marked cellular accumulation, indicating an expanded potential for target inhibition. The concentrations correlated with sensitivity: for sunitinib from 1.5 mM for the most sensitive PDTO to 0.72 mM for the least sensitive PDTO. Likewise, the corresponding cediranib concentrations were 0.15 mM vs 0.062 mM. All PDTOs had high intra-tumoroid osimertinib concentrations (0.90-1.6 mM). Lastly, we detected a significant increase in apoptosis after 3h of HDSE with osimertinib. Whereas our HDSE regimen shows promising results for all 3 TKIs, very short exposure with high-dose osimertinib effectively reduces proliferation and induces cell death in all mCRC PDTOs. While this is likely due to high intra-tumoroid concentrations reached by osimertinib—the mechanism of action at these concentrations remains unknown and is subject to further studies. In parallel, we propose that a HDSE osimertinib regimen warrants clinical exploration as a potential new treatment option for mCRC. Citation Format: Kirti K. Iyer, Dennis Poel, Anne Miggelenbrink, Wouter Kerkhof, Erik van den Hombergh, Loek A. de Jong, Nielka P. van Erp, Daniele V. Tauriello, Henk M. Verheul. High-dose short-exposure of osimertinib robustly inhibits growth of patient-derived metastatic colorectal cancer organoids. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4028.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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 ...
  • 5
    Online Resource
    Online Resource
    MDPI AG ; 2020
    In:  Cancers Vol. 12, No. 7 ( 2020-07-12), p. 1875-
    In: Cancers, MDPI AG, Vol. 12, No. 7 ( 2020-07-12), p. 1875-
    Abstract: Colorectal cancer (CRC) is one of the most frequent and deadly forms of cancer. About half of patients are affected by metastasis, with the cancer spreading to e.g., liver, lungs or the peritoneum. The majority of these patients cannot be cured despite steady advances in treatment options. Immunotherapies are currently not widely applicable for this disease, yet show potential in preclinical models and clinical translation. The tumour microenvironment (TME) has emerged as a key factor in CRC metastasis, including by means of immune evasion—forming a major barrier to effective immuno-oncology. Several approaches are in development that aim to overcome the immunosuppressive environment and boost anti-tumour immunity. Among them are vaccination strategies, cellular transplantation therapies, and targeted treatments. Given the complexity of the system, we argue for rational design of combinatorial therapies and consider the implications of precision medicine in this context.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2020
    detail.hit.zdb_id: 2527080-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 14 ( 2023-1-25)
    Abstract: Colorectal cancer (CRC) remains one of the most aggressive and lethal cancers, with metastasis accounting for most deaths. As such, there is an unmet need for improved therapies for metastatic CRC (mCRC). Currently, the research focus is shifting towards the reciprocal interactions within the tumor microenvironment (TME), which prevent tumor clearance by the immune system. Dendritic cells (DCs) play a key role in the initiation and amplification of anti-tumor immune responses and in driving the clinical success of immunotherapies. Dissecting the interactions between DCs and CRC cells may open doors to identifying key mediators in tumor progression, and possible therapeutic targets. This requires representative, robust and versatile models and tools. Currently, there is a shortage of such in vitro systems to model the CRC TME and its tumor-immune cell interactions. Here we develop and establish a dynamic organotypic 3D co-culture system to recapitulate and untangle the interactions between DCs and patient-derived mCRC tumor organoids. To our knowledge, this is the first study investigating human DCs in co-culture with tumor organoids in a 3D, organotypic setting. This system reveals how mCRC organoids modulate and shape monocyte-derived DCs (MoDCs) behavior, phenotype, and function, within a collagen matrix, using techniques such as brightfield and fluorescence microscopy, flow cytometry, and fluorescence-activated cell sorting. Our 3D co-culture model shows high viability and extensive interaction between DCs and tumor organoids, and its structure resembles patient tissue sections. Furthermore, it is possible to retrieve DCs from the co-cultures and characterize their phenotypic and functional profile. In our study, the expression of activation markers in both mature and immature DCs and their ability to activate T cells were impacted by co-culture with tumor organoids. In the future, this direct co-culture platform can be adapted and exploited to study the CRC-DC interplay in more detail, enabling novel and broader insights into CRC-driven DC (dys)function.
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2023
    detail.hit.zdb_id: 2606827-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for the Advancement of Science (AAAS) ; 2019
    In:  Science Vol. 363, No. 6431 ( 2019-03-08), p. 1051-1051
    In: Science, American Association for the Advancement of Science (AAAS), Vol. 363, No. 6431 ( 2019-03-08), p. 1051-1051
    Abstract: Over the past several decades, evidence has accumulated that stromal cells—the nonmutated cells that surround malignant cancer cells—are not mere bystanders in tumorigenesis. Instead, they play a crucial role in cancer progression. We now know that metastatic cancer cells must reprogram the tumor stroma, or tumor microenvironment (TME), before they can metastasize ( 1 – 3 ). They must also contend with the immune system that strives to limit tumor spreading. If the road to metastasis is full of intense obstacles, how can this disease be so devastating?
    Type of Medium: Online Resource
    ISSN: 0036-8075 , 1095-9203
    RVK:
    RVK:
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2019
    detail.hit.zdb_id: 128410-1
    detail.hit.zdb_id: 2066996-3
    detail.hit.zdb_id: 2060783-0
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 109, No. 14 ( 2012-04-03)
    Abstract: Taken together, these findings significantly change the current view of how Fz relays its signals to Dvl, giving the Fz third loop region and the Dvl DEP-C region a central role. These results provide a mechanistic explanation for a number of functionally defective Drosophila fz gene types, in which mutated residues map to the newly identified third loop Dvl binding motifs ( 3 , 4 ). The previously undescribed Fz-Dvl binding mode implicates a conformation-sensitive interface, which offers possibilities for regulation of the interaction by allosteric means. Understanding how Fz and Dvl interact will help guide the ongoing design of therapeutics that interfere with Wnt pathway activation in cancer. What part of Dvl binds the newly identified motifs in the third intracellular loop of Fz? We confirmed that the Dvl PDZ domain binds the classic KTxxxW motif in vitro, but no interactions were detected with the Fz third loop. Screening of multiple Dvl fragments against the Fz peptide library revealed that, unexpectedly, the Dvl DEP domain plus C terminus (DEP-C) fully recapitulated binding to all three motifs in the Fz receptor. Indeed, the isolated DEP-C domain, but not the PDZ domain, is efficiently recruited to form stable complexes with Fz at the plasma membrane in cells. These findings strongly suggest that the DEP-C domain comprises the Fz binding module in the Dvl protein. Our results further show that the DEP domain mainly contacts the classic motif in the Fz C-tail, whereas the Dvl C-terminal domain binds the third intracellular loop of Fz. Mutational analysis suggests that the very 13 C-terminal amino acids of the Dvl protein comprise a previously unrecognized small functional unit, which helps to stabilize binding of Dvl to the Fz receptor. In this work, we shed light on this issue by using a combinatorial peptide library, which mimics the cytoplasmic interface of the Fz receptor and allows for the identification of complex molecular-recognition modes. We show that Dvl binds a three-segmented, discontinuous site in Fz composed of two previously undescribed motifs in the third intracellular loop and the classic KTxxxW motif in the C-terminal tail ( Fig. P1 ). Furthermore, we demonstrate the functional importance of the highly conserved third loop motifs in Dvl binding and signaling in the context of full-length Fz proteins in human cells and Xenopus embryos. Mutations of single amino acids within these motifs but not in flanking residues disrupt Dvl binding and signaling, suggesting that each of the individual motifs is required but not sufficient for Fz–Dvl complex formation and signal relay. Currently, no Fz receptor crystal structure is available; however, based on homology with related proteins, we show that the three identified motifs are likely to compose a single binding surface on a favorable conformation of the receptor in the membrane ( Fig. P1 ). Wnt binds to receptors on the cell surface called Frizzled (Fz) proteins, which then recruit another protein, Dishevelled (Dvl), usually located in the cell's cytoplasm, to the plasma membrane. Fz–Dvl complexes can then activate signaling cascades that control cellular developmental processes by affecting gene activity. Fz proteins traverse the lipid bilayer seven times, exposing three loops and an extended C-terminal tail in the cytoplasm ( Fig. P1 ). Over a decade ago, a conserved linear motif was identified in the Fz C-terminal tail (KTxxxW) that mediates membrane relocalization of Dvl and activation of Wnt signaling through the β-catenin pathway ( 1 ). Subsequently, a protein interaction module in Dvl, the PDZ domain, was shown to bind the KTxxxW sequence in vitro, and microinjection of Fz peptides comprising this motif were found to interfere with Wnt/β-catenin signaling, confirming the functional importance of the Fz C-terminal tail ( 2 ). Despite this progress, insight into how the interaction between Fz and Dvl drives pathway activation has been scarce. Wnt proteins control embryonic development and are implicated in tumorigenesis. The complex molecular mechanisms mediating Wnt-induced cell responses are of wide interest, and the underlying protein interactions provide an attractive drug target, particularly in cancer treatment. However, the initiating step by which Wnts steer cellular signaling cascades and, ultimately, drive gene activation remains a major unresolved question. Here, we provide key mechanistic insight into how Wnt forms stable protein signaling complexes at the plasma membrane to induce downstream gene activation.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2012
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Nature, Springer Science and Business Media LLC, Vol. 611, No. 7936 ( 2022-11-17), p. 603-613
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Nature, Springer Science and Business Media LLC, Vol. 530, No. 7590 ( 2016-2), p. 340-343
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2016
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    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