Journal articles on the topic 'Tumour organoids'

To see the other types of publications on this topic, follow the link: Tumour organoids.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Tumour organoids.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Munro, Matthew J., Swee T. Tan, and Clint Gray. "Applications for Colon Organoid Models in Cancer Research." Organoids 2, no. 1 (January 12, 2023): 37–49. http://dx.doi.org/10.3390/organoids2010003.

Full text
Abstract:
Organoids are 3D organ-like structures grown from stem cells in vitro that mimic the organ or disease from which they are derived. Due to their stem cell origin, organoids contain a heterogeneous population of cells reflecting the diversity of cell types seen in vivo. Similarly, tumour organoids reflect intratumoural heterogeneity in a way that traditional 2D cell culture and cell lines do not, and, therefore, they show greater promise as a more relevant model for effective disease modelling and drug testing. Tumour organoids arise from cancer stem cells, which contribute to many of the greatest challenges to cancer treatment, including therapy resistance, tumour recurrence, and metastasis. In this review, we outline methods for generating colon organoids from patient-derived normal and tumour tissues. Furthermore, we discuss organoid biobanking, applications of organoids in disease modelling, and a range of platforms applicable to high-throughput drug testing, including apical-out/reverse-polarity colon organoids.
APA, Harvard, Vancouver, ISO, and other styles
2

LeBlanc, VG, D. Trinh, M. Hughes, I. Luthra, D. Livingstone, MD Blough, JG Cairncross, JJ Kelly, and MA Marra. "1450-1545 Young Investigator Awards & Presentations Basic/Translational Exploring cellular subpopulations in glioblastoma and matched organoids using single-cell RNA-seq 52." Canadian Journal of Neurological Sciences / Journal Canadien des Sciences Neurologiques 45, S3 (June 2018): S13—S14. http://dx.doi.org/10.1017/cjn.2018.297.

Full text
Abstract:
Glioblastomas (GBMs) account for nearly half of all primary malignant brain tumours, and current therapies are often only marginally effective. Our understanding of the underlying biology of these tumours and the development of new therapies have been complicated in part by widespread inter- and intratumoural heterogeneity. To characterize this heterogeneity, we performed regional subsampling of primary glioblastomas and derived organoids from these tissue samples. We then performed single-cell RNA-sequencing (scRNA-seq) on these primary regional subsamples and 1-3 matched organoids per sample. We have profiled samples from six tumour sets to date and have obtained sequencing data for 21,234 primary tissue cells and 14,742 organoid cells. While the most apparent differences in gene expression appear to be between individual tumours, we were also able to identify similar cellular subpopulations across tissue samples and across organoids. Importantly, organoids derived from the same tissue sample appeared to be composed of similar cellular subpopulations and were highly comparable to each other, indicating that replicate organoids faithfully represent the original tumour tissue. Overall, our scRNA-seq approach will help evaluate the utility of tumour-derived organoids as model systems for GBM and will aid in identifying cellular subpopulations defined by gene expression patterns, both in primary GBM regional subsamples and their associated organoids. These analyses will allow for the characterization of clonal or subclonal populations that are likely to respond to different therapeutic approaches and may also uncover novel therapeutic targets previously unrevealed through bulk analyses.
APA, Harvard, Vancouver, ISO, and other styles
3

Pinho, Diana, Denis Santos, Ana Vila, and Sandra Carvalho. "Establishment of Colorectal Cancer Organoids in Microfluidic-Based System." Micromachines 12, no. 5 (April 28, 2021): 497. http://dx.doi.org/10.3390/mi12050497.

Full text
Abstract:
Colorectal cancer is the second leading cause of cancer death worldwide. Significant advances in the molecular mechanisms underlying colorectal cancer have been made; however, the clinical approval of new drugs faces many challenges. Drug discovery is a lengthy process causing a rapid increase in global health care costs. Patient-derived tumour organoids are considered preclinical models with the potential for preclinical drug screening, prediction of patient outcomes, and guiding optimized therapy strategies at an individual level. Combining microfluidic technology with 3D tumour organoid models to recapitulate tumour organization and in vivo functions led to the development of an appropriate preclinical tumour model, organoid-on-a-chip, paving the way for personalized cancer medicine. Herein, a low-cost microfluidic device suitable for culturing and expanding organoids, OrganoidChip, was developed. Patient-derived colorectal cancer organoids were cultured within OrganoidChip, and their viability and proliferative activity increased significantly. No significant differences were verified in the organoids’ response to 5-fluorouracil (5-FU) treatment on-chip and on-plate. However, the culture within the OrganoidChip led to a significant increase in colorectal cancer organoid-forming efficiency and overall size compared with conventional culture on a 24-well plate. Interestingly, early-stage and late-stage organoids were predominantly observed on-plate and within the OrganoidChip, respectively. The OrganoidChip thus has the potential to generate in vivo-like organotypic structures for disease modelling and drug screening applications.
APA, Harvard, Vancouver, ISO, and other styles
4

M. Kholosy, Waleed, Marc Derieppe, Femke van den Ham, Kim Ober, Yan Su, Lars Custers, Linda Schild, et al. "Neuroblastoma and DIPG Organoid Coculture System for Personalized Assessment of Novel Anticancer Immunotherapies." Journal of Personalized Medicine 11, no. 9 (August 30, 2021): 869. http://dx.doi.org/10.3390/jpm11090869.

Full text
Abstract:
Cancer immunotherapy has transformed the landscape of adult cancer treatment and holds a great promise to treat paediatric malignancies. However, in vitro test coculture systems to evaluate the efficacy of immunotherapies on representative paediatric tumour models are lacking. Here, we describe a detailed procedure for the establishment of an ex vivo test coculture system of paediatric tumour organoids and immune cells that enables assessment of different immunotherapy approaches in paediatric tumour organoids. We provide a step-by-step protocol for an efficient generation of patient-derived diffuse intrinsic pontine glioma (DIPG) and neuroblastoma organoids stably expressing eGFP-ffLuc transgenes using defined serum-free medium. In contrast to the chromium-release assay, the new platform allows for visualization, monitoring and robust quantification of tumour organoid cell cytotoxicity using a non-radioactive assay in real-time. To evaluate the utility of this system for drug testing in the paediatric immuno-oncology field, we tested our in vitro assay using a clinically used immunotherapy strategy for children with high-risk neuroblastoma, dinutuximab (anti-GD2 monoclonal antibody), on GD2 proficient and deficient patient-derived neuroblastoma organoids. We demonstrated the feasibility and sensitivity of our ex vivo coculture system using human immune cells and paediatric tumour organoids as ex vivo tumour models. Our study provides a novel platform for personalized testing of potential anticancer immunotherapies for aggressive paediatric cancers such as neuroblastoma and DIPG.
APA, Harvard, Vancouver, ISO, and other styles
5

van Tienderen, Groot Koerkamp, IJzermans, van der Laan, and Verstegen. "Recreating Tumour Complexity in a Dish: Organoid Models to Study Liver Cancer Cells and their Extracellular Environment." Cancers 11, no. 11 (November 1, 2019): 1706. http://dx.doi.org/10.3390/cancers11111706.

Full text
Abstract:
Primary liver cancer, consisting predominantly of hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), remains one of the most lethal malignancies worldwide. This high malignancy is related to the complex and dynamic interactions between tumour cells, stromal cells and the extracellular environment. Novel in vitro models that can recapitulate the tumour are essential in increasing our understanding of liver cancer. Herein, primary liver cancer-derived organoids have opened up new avenues due to their patient-specificity, self-organizing ability and potential recapitulation of many of the tumour properties. Organoids are solely of epithelial origin, but incorporation into co-culture models can enable the investigation of the cellular component of the tumour microenvironment. However, the extracellular component also plays a vital role in cancer progression and representation is lacking within current in vitro models. In this review, organoid technology is discussed in the context of liver cancer models through comparisons to other cell culture systems. In addition, the role of the tumour extracellular environment in primary liver cancer will be highlighted with an emphasis on its importance in in vitro modelling. Converging novel organoid-based models with models incorporating the native tumour microenvironment could lead to experimental models that can better recapitulate liver tumours in vivo.
APA, Harvard, Vancouver, ISO, and other styles
6

Soto, Carolina Bizama, and Juan Carlos Roa. "Explorations in the development of novel early diagnosis and disease monitoring tools in digestive tract cancers." Impact 2018, no. 3 (June 15, 2018): 22–23. http://dx.doi.org/10.21820/23987073.2018.3.22.

Full text
Abstract:
Exploring the possibility that genomic DNA (exoDNA) may represent a highly promising next-generation biomarker for early detection. The research team hope to validate the use of circulating exoDNA as a new generation biomarker using KRAS mutation status as a surrogate source of primary tumour information. This may be useful for early diagnosis in cases of pancreatic biliary cancer (PBC) and a valid tool for clinical decision making, disease monitoring and therapeutic stratification. Point mutations in KRAS are found in 50% of gallbladder cancers, thus the team will use an ultra-sensitive droplet digital PCR (ddPCR) platform to identify mutant KRAS in samples of circulating exoDNA isolated from 75 early stage pancreatic biliary cancer patients at the time of diagnosis. These samples will be matched with exoDNA from 'benign disease' controls that have been match on the basis of age and gender. Roa hopes to use exoDNA to query the 'actionable' exome in PBC using a head-to-head comparison using next generation sequencing. Bizama's main objective in investigating patient-derived organoids is to establish and characterise organoid cultures from GBC patients in order to evaluate the drug responses of these organoids both to standard treatments and to specific pathway inhibitors. Current human GBC research lacks in vitro models that can capture the pathophysiological traits of the original tumour and normal epithelia. The research team hope to demonstrate that organoid cultures can be successfully established from normal and tumour gallbladder tissues from Chilean patients and that these organoids will be representative of the primary specimens. The team plan to develop an organoid culture platform as a prototype for the prediction of prognosis and efficiency of standard and targeted therapy. This ex vivo organoids model project may help clinicians predict the response of particular patients to specific treatments, thus avoiding unnecessary therapies.
APA, Harvard, Vancouver, ISO, and other styles
7

Quinn, Jean A., Meera Patel, Kathryn AF Pennel, Dustin Flanagan, Paul G. Horgan, Donald C. McMillan, Simon MacKay, Owen Sansom, and Joanne Edwards. "IKKα as a potential novel target for treatment of colorectal cancer." Journal of Clinical Oncology 38, no. 4_suppl (February 1, 2020): 174. http://dx.doi.org/10.1200/jco.2020.38.4_suppl.174.

Full text
Abstract:
174 Background: Colorectal cancer (CRC) is a heterogeneous disease leading to different survival outcomes for patients with the same stage of disease. The non-canonical NF-κB pathway has been shown to have a key role in tumorigenesis, and the aim of this study was to investigate the role of IKKα, the main catalytic component of this pathway in CRC. Methods: A tissue microarray was retrospectively constructed from a patient cohort (1033) with stage I-III CRC who underwent surgery. IHC was utilised to examine cytoplasmic and punctate IKKα expression and determine any association with clincopathological features and cancer specific survival (CSS). To assess IKKα inhibition, organoids were prepared from wild type (WT) mouse colon, mouse models of CRC (Apc and Apc.KRAS.pT53.TGFbR2 (AKPT)) and patient derived human organoids. These were treated with an IKKα inhibitor, SU1433 and organoid size and cell viability assessed. Results: High cytoplasmic expression of IKKα was associated with increasing T stage (p = 0.012), poor tumour differentiation (p = 0.010), tumour necrosis (p = 0.013) and low proliferation status (p = 0.013) but was not associated with CSS. High punctate IKKα expression associated with tumour differentiation (p = 0.001), necrosis (p = 0.004), proliferation (p = 0.044) and MMR competence (p < 0.001) and was also significantly associated with reduced CSS (HR1.20 95%CI 1.02-1.42, p < 0.001). SU1433 did not significantly impact on WT (C57/B16) organoid viability up to a concentration of 1 uM, however organoid size and cell viability was significantly reduced in a dose dependent manner in organoids from both Apc and AKPT mouse models. A similar reduction was observed in patient derived human organoids. Conclusions: Punctate IKKα expression was associated with poor cancer specific survival in CRC patients, and inhibition with SU1433, impacted on CRC mouse and patient derived human organoid size and cell viability. These results suggest that, following further investigation and confirmation, IKKα may be employed as a novel therapeutic target in CRC.
APA, Harvard, Vancouver, ISO, and other styles
8

Roelofs, Charlotte, Frédéric Hollande, Richard Redvers, Robin L. Anderson, and Delphine Merino. "Breast tumour organoids: promising models for the genomic and functional characterisation of breast cancer." Biochemical Society Transactions 47, no. 1 (January 9, 2019): 109–17. http://dx.doi.org/10.1042/bst20180375.

Full text
Abstract:
Abstract Until recently, established cancer cell lines have been used extensively in breast cancer research, due largely to the difficulties associated with the manipulation and long-term maintenance in culture of primary tumour cells from patients. The recent development of organoid cultures has provided new opportunities to model and analyse patient samples, allowing the propagation of malignant cells under conditions that resemble the three-dimensional growth of breast tumours. They have proved efficacious in preserving the heterogeneity of primary samples and are emerging as a new model to further characterise the molecular features of breast cancer. Organoids formed from patient-derived cells are now in use for the evaluation of drug sensitivity and to validate disease-causing genomic variations. Here, the advantages and limitations of organoid cultures will be discussed and compared with the parallel development of other two- and three-dimensional culture strategies and with patient-derived xenografts. In particular, we will focus on the molecular characterisation of breast cancer organoids and provide some examples of how they have been used in functional studies.
APA, Harvard, Vancouver, ISO, and other styles
9

Ramsay, Robert George. "Rapid in vitro evaluation of immune responses to tumor-derived organoids as an adjunct to immunotherapy trials." Journal of Clinical Oncology 35, no. 15_suppl (May 20, 2017): 3573. http://dx.doi.org/10.1200/jco.2017.35.15_suppl.3573.

Full text
Abstract:
3573 Background: Cancer immunotherapy has made rapid advances with the development of agents that subvert the negative arm of the immune system. This has been important because patients can mount anti-tumor immune responses. In the case of colorectal cancer (CRC), the presence of tumour infiltrating lymphocytes (TILs) appears to have predictive power regarding outcome. Nevertheless, assays that directly evaluate the quality, phenotype and anti-tumor activity of TILs are lacking. Here a novel immune assay platform is presented that measures the kinetics of TIL killing which correlates with pathological tumour response after treatment. Methods: Treatment naïve fresh cancer biopsies were processed to generate organoids and TILs from patients (n = 12) with pathological complete response (pCR) verses non-responding tumours (NRT). These were co-cultured with TILs to organoid ratios of 1:1, 5:1 and 10:1 for 48 hs and TILs function were measured by cytokine release and mean fluorescence intensity (MFI) based upon activated caspase activity. Additionally, TILs from patients with metastatic CRC (n = 20) have also been evaluated. Results: Ten thousand+ organoids are routinely cultured while millions of TILs are retrieved and expanded from biopsies. TIL-mediated killing of patient-matched tumor organoid confirm CD8+ve specific killing. At 24hrs MFI was significantly higher in pCR organoid indicative of immune-mediated cell death compared to NRT organoids at a ratio of 1:1. The efficiency of TIL killing was further enhanced as the ratio increased to 5:1 and 10:1. Gamma interferon production by cytotoxic (CD8+) T-cell is a robust measure of TIL activation state and was also significantly higher by pCR TILs compared to NRT TILs. Substantive differences in TIL subsets were also found in mCRC-derived samples compared to primary CRC. Conclusions: This is a novel functional immune assay and the first of its kind that successfully demonstrated the differences in patient-matched TIL killing between non-responding and responding CRC. This assay can be performed within in two weeks and thus it has translational potential that may change clinical management in the future and immunotherapy strategy design.
APA, Harvard, Vancouver, ISO, and other styles
10

Porter, Ross J., Graeme I. Murray, and Mairi H. McLean. "Current concepts in tumour-derived organoids." British Journal of Cancer 123, no. 8 (July 30, 2020): 1209–18. http://dx.doi.org/10.1038/s41416-020-0993-5.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Amato, Francesco, Colin Rae, Maria Giuseppina Prete, and Chiara Braconi. "Cholangiocarcinoma Disease Modelling Through Patients Derived Organoids." Cells 9, no. 4 (March 30, 2020): 832. http://dx.doi.org/10.3390/cells9040832.

Full text
Abstract:
Cancer organoids are 3D phenotypic cultures that can be established from resected or biopsy tumour samples and can be grown as mini tumours in the dish. Flourishing evidence supports the feasibility of patient derived organoids (PDO) from a number of solid tumours. Evidence for cholangiocarcinoma (CCA) PDO is still sparse but growing. CCA PDO lines have been established from resected early stage disease, advanced cancers and highly chemorefractory tumours. Cancer PDO was shown to recapitulate the 3D morphology, genomic landscape and transcriptomic profile of the source counterpart. They proved to be a valued model for drug discovery and sensitivity testing, and they showed to mimic the drug response observed in vivo in the patients. However, PDO lack representation of the intratumour heterogeneity and the tumour-stroma interaction. The efficiency rate of CCA PDO within the three different subtypes, intrahepatic, perihilar and distal, is still to be explored. In this manuscript we will review evidence for CCA PDO highlighting advantages and limitations of this novel disease model.
APA, Harvard, Vancouver, ISO, and other styles
12

Risbridger, Gail Petuna, Laura Helen Porter, Joe Zhu, David Byrne, Natalie Lister, Arun Azad, Michael Hofman, et al. "Preclinical Evidence of the Efficacy of Lewis Y Car T Cells in Patient-Derived Models of Prostate Cancer." Journal of the Endocrine Society 5, Supplement_1 (May 1, 2021): A1029—A1030. http://dx.doi.org/10.1210/jendso/bvab048.2106.

Full text
Abstract:
Abstract Chimeric antigen receptor T (CAR T) cell therapy is an adoptive immunotherapy that has led to new treatments for lymphoma, leukemia, and other blood cancers; however, its efficacy for prostate cancer remains unproven. Here we report pre-clinical evidence of the efficacy of CAR T cell therapy against the Lewis Y antigen (LeY) using patient-derived models of prostate cancer. To assess the expression of LeY on prostate tumours, we performed immunohistochemistry on a cohort of 41 patient-derived xenografts (PDXs). Cytoplasmic and membrane expression were separately assessed and quantified, for each patient. Overall, 61% (25/41) of PDXs were positive for membrane LeY expression, of which 18 PDXs had greater than 50% membrane-positive cells, and considered most suitable to detection and stable binding by anti-LeY CAR T’s. To determine the in vitro sensitivity to CAR T cytotoxicity, we selected 4 PDXs with high and 2 PDXs with low LeY expression using 3 androgen receptor (AR)-positive adenocarcinomas and 3 AR-negative tumors expressing neuroendocrine markers. Next we established organoids for in vitro co-culture assays where organoids were co-incubated with an equal number of anti-LeY+ CAR T cells or Empty vector control CAR T cells (Ev CAR T). Using time-lapse microscopy we reported destruction of organoids by LeY+ CAR T cells as indicated by their morphological collapse and uptake of propidium iodide from the culture medium; control Ev CAR T cells produced no cytotoxicity. Over the 48h assay, the level of target cell death of the LeY+ organoids was correlated to the intensity LeY surface expression. Target cell death mediated by the CAR T cells required perforin and granzyme B, as potent and highly specific small molecule inhibitors of perforin (SN34960) and granzyme B (C20) applied alone or in combination greatly decreased PI uptake, indicating organoid survival. Neither inhibitor adversely affected CAR T cell viability as measured by PI and Annexin V staining. This demonstrated canonical activation of granule exocytosis pathway by the CAR T cells, leading to organoid cell death. To assess CAR T cell efficacy in vivo, we selected one PDX with high LeY expression. Monotherapy with CAR T cells failed to decrease tumour volume compared to vehicle control. However, CAR T cells given after a single dose of the chemotherapeutic agent carboplatin greatly and durably reduced tumour burden, with residual tumour mass being less than 1% of their original size (0.56 ± 0.23% of tumour volume at the start of treatment). Overall, these data provide preclinical evidence that: i) high membrane expression of LeY correlates with in vitro and in vivo CAR T cell-induced tumour cell death via the canonical perforin/granzyme B mechanism; and, ii) membrane LeY can be used as a biomarker for patient selection.
APA, Harvard, Vancouver, ISO, and other styles
13

Scognamiglio, Giosue, Annarosaria De Chiara, Antonina Parafioriti, Elisabetta Armiraglio, Flavio Fazioli, Michele Gallo, Laura Aversa, et al. "Patient-derived organoids as a potential model to predict response to PD-1/PD-L1 checkpoint inhibitors." British Journal of Cancer 121, no. 11 (October 31, 2019): 979–82. http://dx.doi.org/10.1038/s41416-019-0616-1.

Full text
Abstract:
Abstract Selection of cancer patients for treatment with immune checkpoint inhibitors remains a challenge due to tumour heterogeneity and variable biomarker detection. PD-L1 expression in 24 surgical chordoma specimen was determined immunohistochemically with antibodies 28-8 and E1L3N. The ability of patient-derived organoids to detect treatment effects of nivolumab was explored by quantitative and qualitative immunofluorescence and FACS analysis. The more sensitive antibody, E1L3N (ROC = 0.896, p = 0.001), was associated with greater tumour diameters (p = 0.014) and detected both tumour cells and infiltrating lymphocytes in 54% of patients, but only 1–15% of their cells. Organoids generated from PD-L1-positive patients contained both tumour cells and PD-1/CD8-positive lymphocytes and responded to nivolumab treatment with marked dose-dependent diameter reductions of up to 50% and increased cell death in both PD-L1-positive and negative organoids. Patient-derived organoids may be valuable to predict individual responses to immunotherapy even in patients with low or no immunohistochemical PD-L1 expression.
APA, Harvard, Vancouver, ISO, and other styles
14

Rae, Colin, Francesco Amato, and Chiara Braconi. "Patient-Derived Organoids as a Model for Cancer Drug Discovery." International Journal of Molecular Sciences 22, no. 7 (March 27, 2021): 3483. http://dx.doi.org/10.3390/ijms22073483.

Full text
Abstract:
In the search for the ideal model of tumours, the use of three-dimensional in vitro models is advancing rapidly. These are intended to mimic the in vivo properties of the tumours which affect cancer development, progression and drug sensitivity, and take into account cell–cell interactions, adhesion and invasiveness. Importantly, it is hoped that successful recapitulation of the structure and function of the tissue will predict patient response, permitting the development of personalized therapy in a timely manner applicable to the clinic. Furthermore, the use of co-culture systems will allow the role of the tumour microenvironment and tissue–tissue interactions to be taken into account and should lead to more accurate predictions of tumour development and responses to drugs. In this review, the relative merits and limitations of patient-derived organoids will be discussed compared to other in vitro and ex vivo cancer models. We will focus on their use as models for drug testing and personalized therapy and how these may be improved. Developments in technology will also be considered, including the use of microfluidics, 3D bioprinting, cryopreservation and circulating tumour cell-derived organoids. These have the potential to enhance the consistency, accessibility and availability of these models.
APA, Harvard, Vancouver, ISO, and other styles
15

Nunes, M., C. Trombley, D. E. Flôres, G. Wu, C. Curran, Z. Taleb, J. B. Hogenesch, and P. Karpowicz. "A3 THE LOSS OF THE CIRCADIAN CLOCK GENE BMAL1 INCREASES TUMOUR INITIATION IN APCMIN MICE." Journal of the Canadian Association of Gastroenterology 4, Supplement_1 (March 1, 2021): 3–4. http://dx.doi.org/10.1093/jcag/gwab002.002.

Full text
Abstract:
Abstract Background Circadian rhythms are autonomously running 24h cycles in bodily processes. In animals these rhythms are driven by a molecular time keeper known as the circadian clock. The clock is a transcription-translation feedback loop composed of the transcription factors Bmal1 and Clock as well as their repressors Per and Cry. The circadian clock regulates over 40% of the genome rhythmically. Chronic circadian disruption, in the case of shift work, can lead to pathologies including cancer. Colorectal cancer is most frequently initiated through a mutation in the Wnt pathway regulator, Apc. Several studies have attempted to provide a mechanistic link between cancer and circadian clock disruption but the use of mice on mixed genetic backgrounds and poor circadian models have made this link unclear. Aims We aim to determine if the circadian clock plays a role in intestinal tumourigenesis. Methods We crossed the Apcmin mouse strain, a common intestinal tumour model, with Bmal1 mutant mice, which lack a functioning circadian clock. After creating an isogenic strain, we examined the number of tumours in control (Bmal1+/+) and clock dead (Bmal1-/-) animals. We derived organoids, a 3D cell culture method, from Apc+/+; Bmal1+/+ (healthy, clock-live), Apc+/+, Bmal1-/-(healthy, clock-dead), Apcmin; Bmal1+/+(adenoma, clock-live), Apcmin; Bmal1-/- (adenoma, clock-live) mouse ileum and collected every 2h from 24-48h after synchronizing their circadian clock. Collected samples were sent for RNA sequencing and assessed for circadian regulated transcripts. This experiment was followed up by in vitro organoid assays. Results The circadian clock controls 41 genes in the intestinal epithelium, including genes like Tead4 which are known to be important in intestinal biology. There are twofold more tumours in Bmal1-/- mice than their Bmal1+/+ littermates, and Bmal1-/- tumours upregulate Tead4 and Hippo pathway targets and downregulate Wnt pathway targets. Bmal1-/- adenoma organoids show increased self-renewal when compared to Bmal1+/+ adenoma organoids. However, this increase in self-renewal is lost when organoids are treated with inhibitors of the hippo pathway. Conclusions The circadian clock is important in maintaining the health of an organism, and disruption of the clock can lead to many health consequences including cancer. We show for the first time that the circadian clock controls the hippo signaling mediator Tead4. Additionally, we show that the loss of the clock leads to an increase in the number of tumours present in the epithelium which are characterized by an increase in hippo signaling. This research shows the important of considering time of day when studying stem cells during homeostasis and in cancer. Funding Agencies CIHRNSERC
APA, Harvard, Vancouver, ISO, and other styles
16

Stone, Louise. "In the bank: bladder organoids recapitulate original tumour." Nature Reviews Urology 15, no. 7 (May 3, 2018): 397. http://dx.doi.org/10.1038/s41585-018-0019-8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
17

van der Vaart, Jelte, Lynn Bosmans, Hanneke Margo van Santen, Menno R. Vriens, and Hans Clevers. "Generation of Adult Stem Cell Derived Organoid Cultures From Thyroid Follicular Cells." Journal of the Endocrine Society 5, Supplement_1 (May 1, 2021): A850—A851. http://dx.doi.org/10.1210/jendso/bvab048.1736.

Full text
Abstract:
Abstract The thyroid is essential for maintaining systemic homeostasis by regulating thyroid hormone concentrations in the bloodstream. Due to the limited number of representative model systems, there is limited understanding of fundamental thyroid biology as well as thyroid carcinogenesis. To fill the caveats in the understanding of thyroid cell biology, we aimed to develop an adult stem cell-derived three-dimensional (3D) organoid culture system using murine and human thyroid follicular cells (TFCs). We have succeeded to grow such an organoid culture system that harbours the complete machinery of hormone production visualised by the presence of colloid in the lumen and essential transporters and enzymes in a polarised cell layer. Both the established murine as human thyroid organoids express canonical thyroid markers PAX8 and NKX2.1/TTF1. Moreover, the thyroid hormone precursor thyroglobulin is expressed in both cultures to similar levels as in tissue. Extensive characterisation furthermore identifies known and new biological insights in TFC subclassification, subcellular organisation and hormone production using state-of-the art techniques like single cell RNA sequencing, transmission electron microscopy and genome editing. These 3D in vitro cultures allow for a variety of thyroid-related studies including the progression of wild type cells towards cancer. Additionally, due to the success of generating patient-specific tumour organoids of primary differentiated thyroid carcinoma and metastasis, insights in drug resistance and metastases can be identified. In short, this newly developed organoid culture of murine and human wild type TFCs as well as tumour tissue opens up an extensive area of research that will help understand the drivers for growth and development of thyroid (cancer) cells and enable studies upon drug responsiveness.
APA, Harvard, Vancouver, ISO, and other styles
18

Dionellis, Vasilis S., Maxim Norkin, Angeliki Karamichali, Giacomo G. Rossetti, Joerg Huelsken, Paloma Ordonez-Moran, and Thanos D. Halazonetis. "Genomic Instability Profiles at the Single Cell Level in Mouse Colorectal Cancers of Defined Genotypes." Cancers 13, no. 6 (March 12, 2021): 1267. http://dx.doi.org/10.3390/cancers13061267.

Full text
Abstract:
The genomes of many human CRCs have been sequenced, revealing a large number of genetic alterations. However, the molecular mechanisms underlying the accumulation of these alterations are still being debated. In this study, we examined colorectal tumours that developed in mice with Apclox/lox, LSL-KrasG12D, and Tp53lox/lox targetable alleles. Organoids were derived from single cells and the spectrum of mutations was determined by exome sequencing. The number of single nucleotide substitutions (SNSs) correlated with the age of the tumour, but was unaffected by the number of targeted cancer-driver genes. Thus, tumours that expressed mutant Apc, Kras, and Tp53 alleles had as many SNSs as tumours that expressed only mutant Apc. In contrast, the presence of large-scale (>10 Mb) copy number alterations (CNAs) correlated strongly with Tp53 inactivation. Comparison of the SNSs and CNAs present in organoids derived from the same tumour revealed intratumoural heterogeneity consistent with genomic lesions accumulating at significantly higher rates in tumour cells compared to normal cells. The rate of acquisition of SNSs increased from the early stages of cancer development, whereas large-scale CNAs accumulated later, after Tp53 inactivation. Thus, a significant fraction of the genomic instability present in cancer cells cannot be explained by aging processes occurring in normal cells before oncogenic transformation.
APA, Harvard, Vancouver, ISO, and other styles
19

Rico, Karen, Suzann Duan, Ritu L. Pandey, Yuliang Chen, Jayati T. Chakrabarti, Julie Starr, Yana Zavros, et al. "Genome analysis identifies differences in the transcriptional targets of duodenal versus pancreatic neuroendocrine tumours." BMJ Open Gastroenterology 8, no. 1 (November 2021): e000765. http://dx.doi.org/10.1136/bmjgast-2021-000765.

Full text
Abstract:
ObjectiveGastroenteropancreatic neuroendocrine tumours (GEP-NETs) encompass a diverse group of neoplasms that vary in their secretory products and in their location within the gastrointestinal tract. Their prevalence in the USA is increasing among all adult age groups.AimTo identify the possible derivation of GEP-NETs using genome-wide analyses to distinguish small intestinal neuroendocrine tumours, specifically duodenal gastrinomas (DGASTs), from pancreatic neuroendocrine tumours.DesignWhole exome sequencing and RNA-sequencing were performed on surgically resected GEP-NETs (discovery cohort). RNA transcript profiles available in the Gene Expression Omnibus were analysed using R integrated software (validation cohort). Digital spatial profiling (DSP) was used to analyse paraffin-embedded GEP-NETs. Human duodenal organoids were treated with 5 or 10 ng/mL of tumor necrosis factor alpha (TNFα) prior to qPCR and western blot analysis of neuroendocrine cell specification genes.ResultsBoth the discovery and validation cohorts of small intestinal neuroendocrine tumours induced expression of mesenchymal and calcium signalling pathways coincident with a decrease in intestine-specific genes. In particular, calcium-related, smooth muscle and cytoskeletal genes increased in DGASTs, but did not correlate with MEN1 mutation status. Interleukin 17 (IL-17) and tumor necrosis factor alpha (TNFα) signalling pathways were elevated in the DGAST RNA-sequencing. However, DSP analysis confirmed a paucity of immune cells in DGASTs compared with the adjacent tumour-associated Brunner’s glands. Immunofluorescent analysis showed production of these proinflammatory cytokines and phosphorylated signal transducer and activator of transcription 3 (pSTAT3) by the tumours and stroma. Human duodenal organoids treated with TNFα induced neuroendocrine tumour genes, SYP, CHGA and NKX6.3.ConclusionsStromal–epithelial interactions induce proinflammatory cytokines that promote Brunner’s gland reprogramming.
APA, Harvard, Vancouver, ISO, and other styles
20

Taskovska, Milena, Mateja Erdani Kreft, and Tomaz Smrkolj. "Current and innovative approaches in the treatment of non-muscle invasive bladder cancer: the role of transurethral resection of bladder tumor and organoids." Radiology and Oncology 54, no. 2 (May 2, 2020): 135–43. http://dx.doi.org/10.2478/raon-2020-0025.

Full text
Abstract:
AbstractBackgroundBladder cancer is the 7th most common cancer in men. About 75% of all bladder cancer are non-muscle invasive (NMIBC). The golden standard for definite diagnosis and first-line treatment of NMIBC is transurethral resection of bladder tumour (TURB). Historically, the monopolar current was used first, today bipolar current is preferred by most urologists. Following TURB, depending on the tumour grade, additional intravesical chemo- or/and immunotherapy is indicated, in order to prevent recurrence and need for surgical resection. Development of new technologies, molecular and cell biology, enabled scientists to develop organoids – systems of human cells that are cultivated in the laboratory and have characteristics of the tissue from which they were harvested. In the field of urologic cancers, the organoids are used mainly for studying the course of different diseases, however, in the field of bladder cancer the data are scarce.ConclusionsDifferent currents - monopolar and bipolar, have different effect on urothelium, that is important for oncological results and pathohistological interpretation. Specimens of bladder cancer can be used for preparation of organoids that are further used for studying carcinogenesis. Bladder organoids are step towards personalised medicine, especially for testing effectiveness of chemo-/immunotherapeutics.
APA, Harvard, Vancouver, ISO, and other styles
21

Heredia-Soto, Victoria, Andrés Redondo, José Juan Pozo Kreilinger, Virginia Martínez-Marín, Alberto Berjón, and Marta Mendiola. "3D Culture Modelling: An Emerging Approach for Translational Cancer Research in Sarcomas." Current Medicinal Chemistry 27, no. 29 (September 2, 2020): 4778–88. http://dx.doi.org/10.2174/0929867326666191212162102.

Full text
Abstract:
Sarcomas are tumours of mesenchymal origin, which can arise in bone or soft tissues. They are rare but frequently quite aggressive and with a poor outcome. New approaches are needed to characterise these tumours and their resistance mechanisms to current therapies, responsible for tumour recurrence and treatment failure. This review is focused on the potential of three-dimensional (3D) in vitro models, including multicellular tumour spheroids (MCTS) and organoids, and the latest data about their utility for the study on important properties for tumour development. The use of spheroids as a particularly valuable alternative for compound high throughput screening (HTS) in different areas of cancer biology is also discussed, which enables the identification of new therapeutic opportunities in commonly resistant tumours.
APA, Harvard, Vancouver, ISO, and other styles
22

Weth, Freya R., Lifeng Peng, Erin Paterson, Swee T. Tan, and Clint Gray. "Utility of the Cerebral Organoid Glioma ‘GLICO’ Model for Screening Applications." Cells 12, no. 1 (December 30, 2022): 153. http://dx.doi.org/10.3390/cells12010153.

Full text
Abstract:
Glioblastoma, a grade IV astrocytoma, is regarded as the most aggressive primary brain tumour with an overall median survival of 16.0 months following the standard treatment regimen of surgical resection, followed by radiotherapy and chemotherapy with temozolomide. Despite such intensive treatment, the tumour almost invariably recurs. This poor prognosis has most commonly been attributed to the initiation, propagation, and differentiation of cancer stem cells. Despite the unprecedented advances in biomedical research over the last decade, the current in vitro models are limited at preserving the inter- and intra-tumoural heterogeneity of primary tumours. The ability to understand and manipulate complex cancers such as glioblastoma requires disease models to be clinically and translationally relevant and encompass the cellular heterogeneity of such cancers. Therefore, brain cancer research models need to aim to recapitulate glioblastoma stem cell function, whilst remaining amenable for analysis. Fortunately, the recent development of 3D cultures has overcome some of these challenges, and cerebral organoids are emerging as cutting-edge tools in glioblastoma research. The opportunity to generate cerebral organoids via induced pluripotent stem cells, and to perform co-cultures with patient-derived cancer stem cells (GLICO model), has enabled the analysis of cancer development in a context that better mimics brain tissue architecture. In this article, we review the recent literature on the use of patient-derived glioblastoma organoid models and their applicability for drug screening, as well as provide a potential workflow for screening using the GLICO model. The proposed workflow is practical for use in most laboratories with accessible materials and equipment, a good first pass, and no animal work required. This workflow is also amenable for analysis, with separate measures of invasion, growth, and viability.
APA, Harvard, Vancouver, ISO, and other styles
23

van Essen, Max, Peter Kilfeather, Sally Cowley, John Jacob, and Esther Becker. "Modelling SHH-Medulloblastoma Using Cerebellar Organoids." Neuro-Oncology 24, Supplement_4 (October 1, 2022): iv2. http://dx.doi.org/10.1093/neuonc/noac200.005.

Full text
Abstract:
Abstract AIMS Medulloblastoma (MB) comprises four main groups (SHH, WNT, group 3 and group 4), with distinct molecular, clinical and demographic features. These differences suggest opportunities for tailored and improved treatment. Despite its promise, translation into targeted therapy has been slow, limited by the lack of laboratory models that recapitulate patient tumours. Sporadic and inherited forms of SHH-MB are frequently due to mutations in the tumour suppressor gene PTCH1. Here, we aim to exploit human induced pluripotent stem cells (hiPSC) and CRISPR/Cas9 gene editing, to target PTCH1 in a clinically relevant context, thereby establishing an innovative model of MB. METHOD CRISPR/Cas9 mutation of PTCH1 in hiPSCs was followed by their differentiation into cerebellar organoids. Mutant organoids were studied for early consequences on cerebellar development and tumorigenesis. RESULTS PTCH1-mutation resulted in enhanced SHH-pathway activity in cerebellar organoids. Homozygous PTCH1-mutation prevented the formation of cerebellar progenitor cells, closely mimicking the effects of high SHH- signalling on neuronal progenitors in early embryonal development. Heterozygous PTCH1-mutant hiPSCs could be differentiated into cerebellar organoids, displayed increased growth rate and expressed oncogenes specific to SHH-MB, resembling preneoplasia and MB stem cells. CONCLUSION These results show that cerebellar organoids are a promising new model to study early oncogenic events in MB. Our model presents opportunities for the discovery of new targets which could benefit patients with a genetic predisposition to MB in particular. Refined knowledge of early drivers will be invaluable to guide drug selection and improve prospects for clinical translation.
APA, Harvard, Vancouver, ISO, and other styles
24

Engel, Chan, Nickless, Hlavca, Richards, Kerr, Oliva, McMurrick, Jardé, and Abud. "Patient-Derived Colorectal Cancer Organoids Upregulate Revival Stem Cell Marker Genes following Chemotherapeutic Treatment." Journal of Clinical Medicine 9, no. 1 (January 2, 2020): 128. http://dx.doi.org/10.3390/jcm9010128.

Full text
Abstract:
Colorectal cancer stem cells have been proposed to drive disease progression, tumour recurrence and chemoresistance. However, studies ablating leucine rich repeat containing G protein-coupled receptor 5 (LGR5)-positive stem cells have shown that they are rapidly replenished in primary tumours. Following injury in normal tissue, LGR5+ stem cells are replaced by a newly defined, transient population of revival stem cells. We investigated whether markers of the revival stem cell population are present in colorectal tumours and how this signature relates to chemoresistance. We examined the expression of different stem cell markers in a cohort of patient-derived colorectal cancer organoids and correlated expression with sensitivity to 5-fluorouracil (5-FU) treatment. Our findings revealed that there was inter-tumour variability in the expression of stem cell markers. Clusterin (CLU), a marker of the revival stem cell population, was significantly enriched following 5-FU treatment and expression correlated with the level of drug resistance. Patient outcome data revealed that CLU expression is associated with both lower patient survival and an increase in disease recurrence. This suggests that CLU is a marker of drug resistance and may identify cells that drive colorectal cancer progression.
APA, Harvard, Vancouver, ISO, and other styles
25

Antonica, Francesco, Francesca Garilli, Maria Del Mar Gardeazabal Bataller, Lucia Santomaso, and Luca Tiberi. "TMOD-06. MODELLING ADULT AND PAEDIATRIC GLIOBLASTOMA MULTIFORME (GBM) USING A GENE SCREEN-BASED APPROACH IN MICE AND HUMAN IPSC-DERIVED CEREBRAL ORGANOIDS." Neuro-Oncology 21, Supplement_6 (November 2019): vi263. http://dx.doi.org/10.1093/neuonc/noz175.1105.

Full text
Abstract:
Abstract Glioblastoma multiforme (GBM) represent the most devastating form of high-grade glioma (HGG) affecting adults and children. Despite a multi-therapeutic approach consisting in surgery, radio- and chemo-therapy the prognosis remains poor. Several models such as xenograft, animal models and recently organoids, have been developed in order to investigate the physiopathology of GBM. Although several mouse models (where either gain- or loss-of function of genes/pathway found altered in patients induce tumour formation) have been generated many aspects of how the tumour is formed, evolves, infiltrates and recurs after treatments remain unclear. Another challenge is the creation of proper GBM models showing intratumor heterogeneity found in the patient tumour but missing in the animal model generated so far. To overcome the lack of that human tumour characteristic, we decided to generate new model of GBM using a gene screen-based approach in mice. Firstly, we analysed the genes found amplified or mutated in GBM patients; secondly, we over-expressed the candidate genes (combination of multiple genes found to be mutated or amplified in specific GBM patients) in the subventricular zone (SVZ) of P2 mouse brain. Thirdly, we analysed the formation of tumour after 2 months. We over-expressed roughly 50 combinations in newborn mice and found that only 3 successfully led to the formation of lesions positive for proliferation and brain tumour marker (i.e. GFAP). We are currently characterising the tumours by DNA methylation analysis and RNA-seq (for further confirmation of the cancer subtype and intratumor heterogeneity) or investigating the cell of origin of the tumour. Moreover, we are testing the tumorigenicity of specific in GBM-amplified/mutated gene combinations in hiPSC-derived cerebal organoids. Our data suggest that a gene screen-based approach can be used for quickly and easily assaying the tumorigenicity of genes found amplified/mutated in GBM patients as well as the biology behind such complex process.
APA, Harvard, Vancouver, ISO, and other styles
26

Mout, Lisanne, Lisanne F. van Dessel, Jaco Kraan, Anouk C. de Jong, Rui P. L. Neves, Sigrun Erkens-Schulze, Corine M. Beaufort, et al. "Generating human prostate cancer organoids from leukapheresis enriched circulating tumour cells." European Journal of Cancer 150 (June 2021): 179–89. http://dx.doi.org/10.1016/j.ejca.2021.03.023.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Kashfi, Seyed Mohammad Hossein, Sheema Almozyan, Nicholas Jinks, Bon-Kyoung Koo, and Abdolrahman S. Nateri. "Morphological alterations of cultured human colorectal matched tumour and healthy organoids." Oncotarget 9, no. 12 (January 19, 2018): 10572–84. http://dx.doi.org/10.18632/oncotarget.24279.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Raimondi, Giulia, Ana Mato-Berciano, Silvia Pascual-Sabater, Maria Rovira-Rigau, Miriam Cuatrecasas, Constantino Fondevila, Santiago Sánchez-Cabús, et al. "Patient-derived pancreatic tumour organoids identify therapeutic responses to oncolytic adenoviruses." EBioMedicine 56 (June 2020): 102786. http://dx.doi.org/10.1016/j.ebiom.2020.102786.

Full text
APA, Harvard, Vancouver, ISO, and other styles
29

Hohwieler, Meike, Anett Illing, Patrick C. Hermann, Tobias Mayer, Marianne Stockmann, Lukas Perkhofer, Tim Eiseler, et al. "Human pluripotent stem cell-derived acinar/ductal organoids generate human pancreas upon orthotopic transplantation and allow disease modelling." Gut 66, no. 3 (September 15, 2016): 473–86. http://dx.doi.org/10.1136/gutjnl-2016-312423.

Full text
Abstract:
ObjectiveThe generation of acinar and ductal cells from human pluripotent stem cells (PSCs) is a poorly studied process, although various diseases arise from this compartment.DesignWe designed a straightforward approach to direct human PSCs towards pancreatic organoids resembling acinar and ductal progeny.ResultsExtensive phenotyping of the organoids not only shows the appropriate marker profile but also ultrastructural, global gene expression and functional hallmarks of the human pancreas in the dish. Upon orthotopic transplantation into immunodeficient mice, these organoids form normal pancreatic ducts and acinar tissue resembling fetal human pancreas without evidence of tumour formation or transformation. Finally, we implemented this unique phenotyping tool as a model to study the pancreatic facets of cystic fibrosis (CF). For the first time, we provide evidence that in vitro,but also in our xenograft transplantation assay, pancreatic commitment occurs generally unhindered in CF. Importantly, cystic fibrosis transmembrane conductance regulator (CFTR) activation in mutated pancreatic organoids not only mirrors the CF phenotype in functional assays but also at a global expression level. We also conducted a scalable proof-of-concept screen in CF pancreatic organoids using a set of CFTR correctors and activators, and established an mRNA-mediated gene therapy approach in CF organoids.ConclusionsTaken together, our platform provides novel opportunities to model pancreatic disease and development, screen for disease-rescuing agents and to test therapeutic procedures.
APA, Harvard, Vancouver, ISO, and other styles
30

Karolak, Aleksandra, Dmitry A. Markov, Lisa J. McCawley, and Katarzyna A. Rejniak. "Towards personalized computational oncology: from spatial models of tumour spheroids, to organoids, to tissues." Journal of The Royal Society Interface 15, no. 138 (January 2018): 20170703. http://dx.doi.org/10.1098/rsif.2017.0703.

Full text
Abstract:
A main goal of mathematical and computational oncology is to develop quantitative tools to determine the most effective therapies for each individual patient. This involves predicting the right drug to be administered at the right time and at the right dose. Such an approach is known as precision medicine. Mathematical modelling can play an invaluable role in the development of such therapeutic strategies, since it allows for relatively fast, efficient and inexpensive simulations of a large number of treatment schedules in order to find the most effective. This review is a survey of mathematical models that explicitly take into account the spatial architecture of three-dimensional tumours and address tumour development, progression and response to treatments. In particular, we discuss models of epithelial acini, multicellular spheroids, normal and tumour spheroids and organoids, and multi-component tissues. Our intent is to showcase how these in silico models can be applied to patient-specific data to assess which therapeutic strategies will be the most efficient. We also present the concept of virtual clinical trials that integrate standard-of-care patient data, medical imaging, organ-on-chip experiments and computational models to determine personalized medical treatment strategies.
APA, Harvard, Vancouver, ISO, and other styles
31

Powley, Ian R., Meeta Patel, Gareth Miles, Howard Pringle, Lynne Howells, Anne Thomas, Catherine Kettleborough, et al. "Patient-derived explants (PDEs) as a powerful preclinical platform for anti-cancer drug and biomarker discovery." British Journal of Cancer 122, no. 6 (January 2, 2020): 735–44. http://dx.doi.org/10.1038/s41416-019-0672-6.

Full text
Abstract:
AbstractPreclinical models that can accurately predict outcomes in the clinic are much sought after in the field of cancer drug discovery and development. Existing models such as organoids and patient-derived xenografts have many advantages, but they suffer from the drawback of not contextually preserving human tumour architecture. This is a particular problem for the preclinical testing of immunotherapies, as these agents require an intact tumour human-specific microenvironment for them to be effective. In this review, we explore the potential of patient-derived explants (PDEs) for fulfilling this need. PDEs involve the ex vivo culture of fragments of freshly resected human tumours that retain the histological features of original tumours. PDE methodology for anti-cancer drug testing has been in existence for many years, but the platform has not been widely adopted in translational research facilities, despite strong evidence for its clinical predictivity. By modifying PDE endpoint analysis to include the spatial profiling of key biomarkers by using multispectral imaging, we argue that PDEs offer many advantages, including the ability to correlate drug responses with tumour pathology, tumour heterogeneity and changes in the tumour microenvironment. As such, PDEs are a powerful model of choice for cancer drug and biomarker discovery programmes.
APA, Harvard, Vancouver, ISO, and other styles
32

Nagle, Peter W., and Robert P. Coppes. "Current and Future Perspectives of the Use of Organoids in Radiobiology." Cells 9, no. 12 (December 9, 2020): 2649. http://dx.doi.org/10.3390/cells9122649.

Full text
Abstract:
The majority of cancer patients will be treated with radiotherapy, either alone or together with chemotherapy and/or surgery. Optimising the balance between tumour control and the probability of normal tissue side effects is the primary goal of radiation treatment. Therefore, it is imperative to understand the effects that irradiation will have on both normal and cancer tissue. The more classical lab models of immortal cell lines and in vivo animal models have been fundamental to radiobiological studies to date. However, each of these comes with their own limitations and new complementary models are required to fill the gaps left by these traditional models. In this review, we discuss how organoids, three-dimensional tissue-resembling structures derived from tissue-resident, embryonic or induced pluripotent stem cells, overcome the limitations of these models and thus have a growing importance in the field of radiation biology research. The roles of organoids in understanding radiation-induced tissue responses and in moving towards precision medicine are examined. Finally, the limitations of organoids in radiobiology and the steps being made to overcome these limitations are considered.
APA, Harvard, Vancouver, ISO, and other styles
33

Lannagan, Tamsin R. M., Young K. Lee, Tongtong Wang, Jatin Roper, Mark L. Bettington, Lochlan Fennell, Laura Vrbanac, et al. "Genetic editing of colonic organoids provides a molecularly distinct and orthotopic preclinical model of serrated carcinogenesis." Gut 68, no. 4 (April 17, 2018): 684–92. http://dx.doi.org/10.1136/gutjnl-2017-315920.

Full text
Abstract:
ObjectiveSerrated colorectal cancer (CRC) accounts for approximately 25% of cases and includes tumours that are among the most treatment resistant and with worst outcomes. This CRC subtype is associated with activating mutations in the mitogen-activated kinase pathway gene, BRAF, and epigenetic modifications termed the CpG Island Methylator Phenotype, leading to epigenetic silencing of key tumour suppressor genes. It is still not clear which (epi-)genetic changes are most important in neoplastic progression and we begin to address this knowledge gap herein.DesignWe use organoid culture combined with CRISPR/Cas9 genome engineering to sequentially introduce genetic alterations associated with serrated CRC and which regulate the stem cell niche, senescence and DNA mismatch repair.ResultsTargeted biallelic gene alterations were verified by DNA sequencing. Organoid growth in the absence of niche factors was assessed, as well as analysis of downstream molecular pathway activity. Orthotopic engraftment of complex organoid lines, but not BrafV600E alone, quickly generated adenocarcinoma in vivo with serrated features consistent with human disease. Loss of the essential DNA mismatch repair enzyme, Mlh1, led to microsatellite instability. Sphingolipid metabolism genes are differentially regulated in both our mouse models of serrated CRC and human CRC, with key members of this pathway having prognostic significance in the human setting.ConclusionWe generate rapid, complex models of serrated CRC to determine the contribution of specific genetic alterations to carcinogenesis. Analysis of our models alongside patient data has led to the identification of a potential susceptibility for this tumour type.
APA, Harvard, Vancouver, ISO, and other styles
34

Lee, Tet Woo, Amy Lai, Julia K. Harms, Dean C. Singleton, Benjamin D. Dickson, Andrew M. J. Macann, Michael P. Hay, and Stephen M. F. Jamieson. "Patient-Derived Xenograft and Organoid Models for Precision Medicine Targeting of the Tumour Microenvironment in Head and Neck Cancer." Cancers 12, no. 12 (December 12, 2020): 3743. http://dx.doi.org/10.3390/cancers12123743.

Full text
Abstract:
Patient survival from head and neck squamous cell carcinoma (HNSCC), the seventh most common cause of cancer, has not markedly improved in recent years despite the approval of targeted therapies and immunotherapy agents. Precision medicine approaches that seek to individualise therapy through the use of predictive biomarkers and stratification strategies offer opportunities to improve therapeutic success in HNSCC. To enable precision medicine of HNSCC, an understanding of the microenvironment that influences tumour growth and response to therapy is required alongside research tools that recapitulate the features of human tumours. In this review, we highlight the importance of the tumour microenvironment in HNSCC, with a focus on tumour hypoxia, and discuss the fidelity of patient-derived xenograft and organoids for modelling human HNSCC and response to therapy. We describe the benefits of patient-derived models over alternative preclinical models and their limitations in clinical relevance and how these impact their utility in precision medicine in HNSCC for the discovery of new therapeutic agents, as well as predictive biomarkers to identify patients’ most likely to respond to therapy.
APA, Harvard, Vancouver, ISO, and other styles
35

Shree Harini, Karthik, Devaraj Ezhilarasan, and Thangavelu Lakshmi. "Patient derived tumour organoids: An emerging strategy in oral cancer research and therapeutics." Oral Oncology 131 (August 2022): 105954. http://dx.doi.org/10.1016/j.oraloncology.2022.105954.

Full text
APA, Harvard, Vancouver, ISO, and other styles
36

Boot, Gina Faye, Federica Panebianco, John Gallon, Charlotte Kiu Yan Ng, and Salvatore Piscuoglio. "Abstract 3074: (Phospho)proteomics profiling of patient-derived colon cancer organoids for the discovery of putative drug targets." Cancer Research 82, no. 12_Supplement (June 15, 2022): 3074. http://dx.doi.org/10.1158/1538-7445.am2022-3074.

Full text
Abstract:
Abstract Introduction: Colorectal cancer is the second cause of cancer related deaths worldwide, with the poor survival outcomes attributable to the presence of metastases. To tackle this problem, (phospho)proteomics analysis enables valuable insights into changes of protein expression and signalling in cancer that can be perturbed by drugs. To study mechanisms driving metastasis and perform subsequent drug testing, patient derived organoids (PDOs) are in development as preclinical models. PDOs are obtained by 3D culture of tumour tissue ex-vivo, which enables them to retain the heterogeneity and architecture of the tumor source. To identify putative drug targets for colon cancer metastasis, we performed comparative (phospho)proteomics analysis of metastatic colon tissues and their matched PDOs. Methods: (Phospho)proteomics profiling was performed on PDOs generated from 10 patients with colon metastases to the liver, 10 matched tumour tissues and 4 normal colon mucosa. Unsupervised analysis of the (phospho)proteomic landscape was used to identify differentially expressed genes and pathways in colon metastases and their matched PDOs, compared to normal colon mucosa. We focused on phosphosites, proteins and pathways showing consistently altered expression in tumour tissues and PDOs. Kinase-substrate enrichment analysis was used to identify aberrantly activated kinases, based on the abundance of phosphorylation on the substrates. Putative drugs were selected using the consensus expression response to multiple small molecule drugs across cell lines and conditions from the Library of Integrated Network-Based Cellular Signatures (LINCS) database. Drug treatment of drug candidates was performed using the MTT Cell Proliferation Assay. Results: Using the approach described above we identified 103 differentially expressed proteins and 236 phosphosites between tumour tissues and normals, which were also detected in PDOs, with overall high correlation of t statistics (0.6 Spearman’s rho) between tumor tissues and PDOs. MYC-targets, G2M checkpoints and E2F-targets were amongst the top positively enriched pathways in common, and the LINCS analysis identified multi-kinase inhibitor Nintedanib and NFKB pathway-targeting KIN0-260 as putative drugs for upregulated proteins. The kinase CSNK2A1 was overactivated in both groups, pointing towards the use of CK2 inhibitors for drug testing. Treatment at 5 days with Nintedanib and KIN001-260 on the PDOs resulted in significant reduction of cell viability compared with 5-FU. Conclusion: Our study provides evidence that PDOs recapitulate relevant tumor features at the proteomic and phosphoproteomic levels, supporting the utility of this ex-vivo model as a tool for drug sensitivity testing for metastatic colon cancer. Citation Format: Gina Faye Boot, Federica Panebianco, John Gallon, Charlotte Kiu Yan Ng, Salvatore Piscuoglio. (Phospho)proteomics profiling of patient-derived colon cancer organoids for the discovery of putative drug targets [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 3074.
APA, Harvard, Vancouver, ISO, and other styles
37

Diksin, Mohammed, Jonathan Rowlinson, Alexandar Kondrashov, Chris Denning, Jamie Hughes, Tim Constantin-Teodo, Wei Cui, et al. "Characterisation of the invasive tumour niche using astrocyte-glioblastoma organoids and decellularised human brain." Neuro-Oncology 21, Supplement_4 (October 2019): iv7. http://dx.doi.org/10.1093/neuonc/noz167.028.

Full text
Abstract:
Abstract Glioblastoma therapeutic challenges are in considerable part due to myriad survival adaptations and mechanisms, which allow malignant cells to repurpose signalling pathways within discreet microenvironments. These Darwinian adaptations facilitate invasion into brain parenchyma and perivascular space or promote evasion from repressive factors that represent anti-cancer defence mechanisms. We hypothesised that pre-clinical modelling of glioma invasion by recapitulating early events occurring immediately after surgery at the glioblastoma invasive margin, could reveal the cross-talk between malignant cells and the surrounding healthy astrocytes, which facilitates tumour recurrence. We first generated transgenic H1-derived neural stem cells using CRISPR/Cas9-mediated knock-in of the YFP reporter gene under the control of the GFAP promoter. Reproducible ultrahigh-throughput AggreWells™ (19,200 micro-wells per 24-well plate) were used to create astrocyte-glioblastoma organoids, which we term ‘Gliomasphere Matrices’. YFP-labelled astrocytes were co-cultured with 10 treatment-naïve patient-derived cell lines isolated from the 5-aminolevulinic (5ALA)-determined glioblastoma invasive margin. Co-cultures were seeded upon on a sequentially constructed, time-of-flight secondary ion mass spectrometry (ToF-SIMS)-characterised 3D scaffold, composed of decellularised human brain extract with defined PEGDA hydrogel. YFP-astrocytes were purified from each of the 10 Gliomasphere Matrices using fluorescence-activated cell sorting (FACS) after 6- and 10-days co-culture. RNAseq profiling to address both putative astrocytic reprogramming by invasive glioblastoma cells and gene expression changes intrinsic to tumour cells will be discussed in relation to RNAseq data from patient-derived 5ALA FACS-purified glioblastoma invasive margin tissue. This novel multi-faceted model offers a unique opportunity to recapitulate early molecular cross-talk which facilitates glioblastoma recurrence and may be utilised for high-throughput drug screening.
APA, Harvard, Vancouver, ISO, and other styles
38

Slostad, Jessica, Ashiq Masood, April T. Swoboda, and Mia Alyce Levy. "Towards the clinical validity of tumor organoid drug screens: Establishing a framework for organoid disease models." Journal of Clinical Oncology 39, no. 15_suppl (May 20, 2021): e15037-e15037. http://dx.doi.org/10.1200/jco.2021.39.15_suppl.e15037.

Full text
Abstract:
e15037 Background: Despite advances in biomarker-directed cancer therapies to predict tumor response to treatment, precision oncology remains an imprecise science. Tumor organoid drug screens present an opportunity to test multiple potentially effective therapies simultaneously before exposing a patient to treatment toxicities. Preliminary studies have established the analytic validity; however, the clinical validity of the tumor organoid response to a drug to predict the tumor response in patients is unclear. Methods: In order to establish the clinical validity of an organoid drug screen, a clinical disease model should have the following features to enable comparison of the tumor’s clinical response to treatment and the organoid’s response to the same treatment. First, a fresh tissue biopsy (non-bone) needs to be obtained for organoid development prior to the start of systemic treatment. Priority should be given to tumors known to successfully grow organoids. Disease models where the standard of care systemic treatment is a single cytotoxic or targeted agent would best assess correlation between the drug screen and patient response, such as metastatic breast, cervical, or prostate cancer. This would often lead to enrolling patients on later line systemic therapies. Studies should avoid drugs whose mechanism of action leverages the patient metabolism or tumor microenvironment (e.g. immunotherapy, aromatase inhibitors, VEGF inhibitors). Patient should have measurable disease that can be measured clinically, radiographically, or pathologically and using standardized response evaluation criteria (e.g. RECIST). Next generation sequencing would assess genomic concordance between the tumor in the patient and organoid. These studies would determine the feasibility and timeliness of prospectively developing tumor organoids that is sufficient to perform a drug screen. Results: We propose two research models to evaluate the clinical validity of tumor organoid drug screens: a metastatic solid tumor and neoadjuvant solid tumor disease model. Metastatic disease models provide an opportunity to assess response across multiple cancer types at the time of progression and initiation of next line of therapy. The efficacy of chemotherapy can be determined using objective data from radiologic response (RECIST). In neoadjuvant models, pathologic (pCR) and radiologic response can provide objective data for organoid response. These study design features will lay the framework for determining the clinical utility of organoid drug screens. Conclusions: We call for clinical studies assessing the clinical validity of tumor organoid drug screens and determining their concordance with patient response to systemic therapy. Advancements in clinically validated tumor organoids have the potential to fundamentally shift clinical paradigms and improve patient outcomes in cancer treatment.
APA, Harvard, Vancouver, ISO, and other styles
39

Shembrey, Carolyn, Jai Smith, Mélodie Grandin, Nathalia Williams, Hyun-Jung Cho, Christina Mølck, Corina Behrenbruch, et al. "Longitudinal Monitoring of Intra-Tumoural Heterogeneity Using Optical Barcoding of Patient-Derived Colorectal Tumour Models." Cancers 14, no. 3 (January 24, 2022): 581. http://dx.doi.org/10.3390/cancers14030581.

Full text
Abstract:
Geno- and phenotypic heterogeneity amongst cancer cell subpopulations are established drivers of treatment resistance and tumour recurrence. However, due to the technical difficulty associated with studying such intra-tumoural heterogeneity, this phenomenon is seldom interrogated in conventional cell culture models. Here, we employ a fluorescent lineage technique termed “optical barcoding” (OBC) to perform simultaneous longitudinal tracking of spatio-temporal fate in 64 patient-derived colorectal cancer subclones. To do so, patient-derived cancer cell lines and organoids were labelled with discrete combinations of reporter constructs, stably integrated into the genome and thus passed on from the founder cell to all its clonal descendants. This strategy enables the longitudinal monitoring of individual cell lineages based upon their unique optical barcodes. By designing a novel panel of six fluorescent proteins, the maximum theoretical subpopulation resolution of 64 discriminable subpopulations was achieved, greatly improving throughput compared with previous studies. We demonstrate that all subpopulations can be purified from complex clonal mixtures via flow cytometry, permitting the downstream isolation and analysis of any lineages of interest. Moreover, we outline an optimized imaging protocol that can be used to image optical barcodes in real-time, allowing for clonal dynamics to be resolved in live cells. In contrast with the limited intra-tumour heterogeneity observed in conventional 2D cell lines, the OBC technique was successfully used to quantify dynamic clonal expansions and contractions in 3D patient-derived organoids, which were previously demonstrated to better recapitulate the heterogeneity of their parental tumour material. In summary, we present OBC as a user-friendly, inexpensive, and high-throughput technique for monitoring intra-tumoural heterogeneity in in vitro cell culture models.
APA, Harvard, Vancouver, ISO, and other styles
40

Usui, Tatsuya, Masashi Sakurai, Shuhei Enjoji, Hideyoshi Kawasaki, Koji Umata, Takashi Ohama, Nobuyuki Fujiwara, et al. "Establishment of a Novel Model for Anticancer Drug Resistance in Three-Dimensional Primary Culture of Tumor Microenvironment." Stem Cells International 2016 (2016): 1–10. http://dx.doi.org/10.1155/2016/7053872.

Full text
Abstract:
Tumor microenvironment has been implicated in tumor development and progression. As a three-dimensional tumor microenvironment model, air liquid interface (ALI) organoid culture from oncogene transgenic mouse gastrointestinal tissues was recently produced. However, ALI organoid culture system from tissues of colorectal cancer patients has not been established. Here, we developed an ALI organoid model from normal and tumor colorectal tissues of human patients. Both organoids were successfully generated and showed cystic structures containing an epithelial layer and surrounding mesenchymal stromal cells. Structures of tumor organoids closely resembled primary tumor epithelium. Expression of an epithelial cell marker, E-cadherin, a goblet cell marker, MUC2, and a fibroblast marker, vimentin, but not a myofibroblast marker, α-smooth muscle actin (SMA), was observed in normal organoids. Expression of E-cadherin, MUC2, vimentin, and α-SMA was observed in tumor organoids. Expression of a cancer stem cell marker, LGR5 in tumor organoids, was higher than that in primary tumor tissues. Tumor organoids were more resistant to toxicity of 5-fluorouracil and Irinotecan than colorectal cancer cell lines, SW480, SW620, and HCT116. These findings indicate that ALI organoid culture from colorectal cancer patients may become a novel model that is useful for examining resistance to chemotherapy in tumor microenvironment.
APA, Harvard, Vancouver, ISO, and other styles
41

Wykes, Victoria, Alina Finch, Daniel Blakeway, Chiara Bardella, Ute Pohl, Andrew Beggs, and Colin Watts. "BRAIN Surgical Tissue for Advanced Tumour models in Precision medicine: Developing the BRAIN-STAT pathway." Neuro-Oncology 23, Supplement_4 (October 1, 2021): iv19—iv20. http://dx.doi.org/10.1093/neuonc/noab195.050.

Full text
Abstract:
Abstract Aims There are approximately four thousand neuro-oncology procedures in the UK per annum. Many of these result in tissue and biofluid specimens that are surplus to diagnostic requirement and can be collected as standard of clinical care. However, developing technologies and treatments for precision medicine require access to a range of individualised biospecimens paired with deep clinical phenotyping data. Here, we present Brain Surgical Tissue for Advanced Tumour Models (BRAINSTAT) programme, an infrastructure that has been established between Queen Elizabeth Hospital, Birmingham and the University of Birmingham, to collect, structure and store these resources and also maximise their value for research over the long-term. Using this approach our aim is to provide high-quality, annotated resources to help develop novel treatments for patients with brain tumours. Method BRAINSTAT infrastructure allows: Prospective consent Biospecimens, including tumour tissue (brain and other primary in the case of metastasis), cyst fluid, dura, skin, CSF, blood (matched “germ-line” and for circulating cell free tumour DNA analysis), urine and saliva can be collected. Consent for long term follow-up, is either via clinic or NHS digital. More limited consent for non-oncological neurosurgical cohorts (e.g. epilepsy or vascular) and healthy volunteers allow healthy access-tissue and biofluids to be collected. B. Rapid transfer of fresh surgical tissue samples: Strong collaborative links and close physical proximity between operating theatre and laboratory allows rapid transfer of biospecimens minimising transit time. C. Standardised annotation across disciplines The RedCAP database system allows granular control over data-access, and each specialist research team is provided access only to the sub-sections relevant to them. All users must have Good Clinical Practice certification and GDPR training, prior to access of the BRAINSTAT database. Results Between 25/11/2019-16/03/2020 and 27/07/2020-16/11/2020, 65 patients were consented for BRAINSTAT at the weekly neurosurgical oncology clinic. (Recruitment gaps due to the SARS-COVID 19 pandemic). Pathological diagnosis of surplus tissue collected included: 37 high grade glioma, 3 low grade glioma and 16 brain metastasis including: (6 lung, 6 breast, 2 colorectal, 1 oesophageal, 1 endometrial). Meningioma (5 WHO I; 1 WHO III) 1 patient undergoing anterior temporal lobectomy for hippocampal sclerosis contributed access tissue from the lateral neocortex. 1 patient had a non- neoplastic, non-diagnostic sample. All patients had matched “germ-line” blood samples. Median time from resection to arrival in the laboratory was 10 minutes (range 4-31). Standardised operating protocols to optimise this have been developed. Glioblastoma and breast-brain metastasis tumourspheres and cerebral organoids are currently being validated. Conclusion Despite the challenges of the pandemic we have established a viable tissue pipeline from neurosurgical operating theatre to our university laboratories. We are developing clinically annotated human brain tumour cell lines, stem cells and 3D organoid models, principally for commonly encountered brain tumours such as glioma and metastasis. The research sets the foundation for a multitude of downstream applications including: - Building more complex organoid cultures e.g. by including other cell types such as healthy brain cells and endothelial cells allowing future experiments to more accurately model tumour growth. - Developing high-throughput, patient-specific drug screens of novel drugs and drug combinations using these 3D tumour models aiming to more effectively treat tumour proliferation and spread. These patient avatars will help inform and test more “stratified” personal medical treatments and will provide opportunities to allow earlier intervention with the aim of improving survival, coupled with a better quality of life.
APA, Harvard, Vancouver, ISO, and other styles
42

Hakuno, Sarah K., Ellis Michiels, Eleonore B. Kuhlemaijer, Ilse Rooman, Lukas J. A. C. Hawinkels, and Marije Slingerland. "Multicellular Modelling of Difficult-to-Treat Gastrointestinal Cancers: Current Possibilities and Challenges." International Journal of Molecular Sciences 23, no. 6 (March 15, 2022): 3147. http://dx.doi.org/10.3390/ijms23063147.

Full text
Abstract:
Cancers affecting the gastrointestinal system are highly prevalent and their incidence is still increasing. Among them, gastric and pancreatic cancers have a dismal prognosis (survival of 5–20%) and are defined as difficult-to-treat cancers. This reflects the urge for novel therapeutic targets and aims for personalised therapies. As a prerequisite for identifying targets and test therapeutic interventions, the development of well-established, translational and reliable preclinical research models is instrumental. This review discusses the development, advantages and limitations of both patient-derived organoids (PDO) and patient-derived xenografts (PDX) for gastric and pancreatic ductal adenocarcinoma (PDAC). First and next generation multicellular PDO/PDX models are believed to faithfully generate a patient-specific avatar in a preclinical setting, opening novel therapeutic directions for these difficult-to-treat cancers. Excitingly, future opportunities such as PDO co-cultures with immune or stromal cells, organoid-on-a-chip models and humanised PDXs are the basis of a completely new area, offering close-to-human models. These tools can be exploited to understand cancer heterogeneity, which is indispensable to pave the way towards more tumour-specific therapies and, with that, better survival for patients.
APA, Harvard, Vancouver, ISO, and other styles
43

van Tienderen, Gilles, Kathryn Monfils, Floris Roos, Luc J. W. van der Laan, and Monique M. A. Verstegen. "Induction of branching morphogenesis in cholangiocarcinoma organoids in vitro improves similarity with the original tumour." Journal of Hepatology 77 (July 2022): S46—S47. http://dx.doi.org/10.1016/s0168-8278(22)00503-7.

Full text
APA, Harvard, Vancouver, ISO, and other styles
44

Calandrini, C., S. Derakhshan, E. M. Dolman, R. De Krijger, L. Tytgat, J. Molenaar, M. Van den Heuvel-Eibrink, and J. Drost. "41P Identifying novel target therapies for paediatric kidney cancer patients using patient-derived tumour organoids." Annals of Oncology 31 (March 2020): S18. http://dx.doi.org/10.1016/j.annonc.2020.01.022.

Full text
APA, Harvard, Vancouver, ISO, and other styles
45

Gunti, Sreenivasulu, Austin T. K. Hoke, Kenny P. Vu, and Nyall R. London. "Organoid and Spheroid Tumor Models: Techniques and Applications." Cancers 13, no. 4 (February 19, 2021): 874. http://dx.doi.org/10.3390/cancers13040874.

Full text
Abstract:
Techniques to develop three-dimensional cell culture models are rapidly expanding to bridge the gap between conventional cell culture and animal models. Organoid and spheroid cultures have distinct and overlapping purposes and differ in cellular sources and protocol for establishment. Spheroids are of lower complexity structurally but are simple and popular models for drug screening. Organoids histologically and genetically resemble the original tumor from which they were derived. Ease of generation, ability for long-term culture and cryopreservation make organoids suitable for a wide range of applications. Organoids-on-chip models combine organoid methods with powerful designing and fabrication of micro-chip technology. Organoid-chip models can emulate the dynamic microenvironment of tumor pathophysiology as well as tissue–tissue interactions. In this review, we outline different tumor spheroid and organoid models and techniques to establish them. We also discuss the recent advances and applications of tumor organoids with an emphasis on tumor modeling, drug screening, personalized medicine and immunotherapy.
APA, Harvard, Vancouver, ISO, and other styles
46

De Santis, Ilaria, Ervin Tasnadi, Peter Horvath, Alessandro Bevilacqua, and Filippo Piccinini. "Open-Source Tools for Volume Estimation of 3D Multicellular Aggregates." Applied Sciences 9, no. 8 (April 18, 2019): 1616. http://dx.doi.org/10.3390/app9081616.

Full text
Abstract:
The volume is one of the most relevant features that define the treatment of an in vivo tumour. When using cancer 3D in vitro models in pre-clinical studies, it becomes important to evaluate the macroscopic effects of drugs and radiotherapy treatments. Depending on the nature of the 3D in vitro model used, different open-source solutions can be used for measuring the volume by starting from microscope-acquired images. In this work, we introduced several open-source tools today available for estimating the volume of 3D multicellular aggregates (e.g., spheroids, organoids), also giving hints for defining the “best software” by analysing characteristics of 3D in vitro models and limits of the tools. Finally, using several cancer organoids imaged by a fluorescent microscope, we compared volume estimations obtained with different tools, besides presenting a new version of the Reconstruction and Visualization from Multiple Sections (ReViMS version 2.0) tool. This work aims to be the reference for researchers interested in estimating the volume of 3D multicellular aggregates through an open-source tool.
APA, Harvard, Vancouver, ISO, and other styles
47

Weeber, Fleur, Marc van de Wetering, Marlous Hoogstraat, Krijn K. Dijkstra, Oscar Krijgsman, Thomas Kuilman, Christa G. M. Gadellaa-van Hooijdonk, et al. "Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases." Proceedings of the National Academy of Sciences 112, no. 43 (October 12, 2015): 13308–11. http://dx.doi.org/10.1073/pnas.1516689112.

Full text
Abstract:
Tumor organoids are 3D cultures of cancer cells. They can be derived from the tumor of each individual patient, thereby providing an attractive ex vivo assay to tailor treatment. Using patient-derived tumor organoids for this purpose requires that organoids derived from biopsies maintain the genetic diversity of the in vivo tumor. In this study tumor biopsies were obtained from 14 patients with metastatic colorectal cancer (i) to test the feasibility of organoid culture from metastatic biopsy specimens and (ii) to compare the genetic diversity of patient-derived tumor organoids and the original tumor biopsy. Genetic analysis was performed using SOLiD sequencing for 1,977 cancer-relevant genes. Copy number profiles were generated from sequencing data using CopywriteR. Here we demonstrate that organoid cultures can be established from tumor biopsies of patients with metastatic colorectal cancer with a success rate of 71%. Genetic analysis showed that organoids reflect the metastasis from which they were derived. Ninety percent of somatic mutations were shared between organoids and biopsies from the same patient, and the DNA copy number profiles of organoids and the corresponding original tumor show a correlation of 0.89. Most importantly, none of the mutations that were found exclusively in either the tumor or organoid culture are in driver genes or genes amenable for drug targeting. These findings support further exploration of patient-derived organoids as an ex vivo platform to personalize anticancer treatment.
APA, Harvard, Vancouver, ISO, and other styles
48

Rottenberg, S., A. Duarte, E. Duarte, P. Francica, M. Mutlu, S. Blatter, N. Sachs, H. Clevers, and J. Jonkers. "BRCA-deficient mouse mammary tumour organoids as a rapid tool to study anti-cancer drug resistance." Journal of Comparative Pathology 166 (January 2019): 120. http://dx.doi.org/10.1016/j.jcpa.2018.10.065.

Full text
APA, Harvard, Vancouver, ISO, and other styles
49

Krastev, Dragomir B., Shudong Li, Yilun Sun, Andrew J. Wicks, Gwendoline Hoslett, Daniel Weekes, Luned M. Badder, et al. "The ubiquitin-dependent ATPase p97 removes cytotoxic trapped PARP1 from chromatin." Nature Cell Biology 24, no. 1 (January 2022): 62–73. http://dx.doi.org/10.1038/s41556-021-00807-6.

Full text
Abstract:
AbstractPoly (ADP-ribose) polymerase (PARP) inhibitors elicit antitumour activity in homologous recombination-defective cancers by trapping PARP1 in a chromatin-bound state. How cells process trapped PARP1 remains unclear. Using wild-type and a trapping-deficient PARP1 mutant combined with rapid immunoprecipitation mass spectrometry of endogenous proteins and Apex2 proximity labelling, we delineated mass spectrometry-based interactomes of trapped and non-trapped PARP1. These analyses identified an interaction between trapped PARP1 and the ubiquitin-regulated p97 ATPase/segregase. We found that following trapping, PARP1 is SUMOylated by PIAS4 and subsequently ubiquitylated by the SUMO-targeted E3 ubiquitin ligase RNF4, events that promote recruitment of p97 and removal of trapped PARP1 from chromatin. Small-molecule p97-complex inhibitors, including a metabolite of the clinically used drug disulfiram (CuET), prolonged PARP1 trapping and enhanced PARP inhibitor-induced cytotoxicity in homologous recombination-defective tumour cells and patient-derived tumour organoids. Together, these results suggest that p97 ATPase plays a key role in the processing of trapped PARP1 and the response of tumour cells to PARP inhibitors.
APA, Harvard, Vancouver, ISO, and other styles
50

Dhakal, Bimala, Celine Man Ying Li, Runhao Li, Kenny Yeo, Josephine A. Wright, Krystyna A. Gieniec, Laura Vrbanac, et al. "The Antianginal Drug Perhexiline Displays Cytotoxicity against Colorectal Cancer Cells In Vitro: A Potential for Drug Repurposing." Cancers 14, no. 4 (February 18, 2022): 1043. http://dx.doi.org/10.3390/cancers14041043.

Full text
Abstract:
Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide. Perhexiline, a prophylactic anti-anginal drug, has been reported to have anti-tumour effects both in vitro and in vivo. Perhexiline as used clinically is a 50:50 racemic mixture ((R)-P) of (−) and (+) enantiomers. It is not known if the enantiomers differ in terms of their effects on cancer. In this study, we examined the cytotoxic capacity of perhexiline and its enantiomers ((−)-P and (+)-P) on CRC cell lines, grown as monolayers or spheroids, and patient-derived organoids. Treatment of CRC cell lines with (R)-P, (−)-P or (+)-P reduced cell viability, with IC50 values of ~4 µM. Treatment was associated with an increase in annexin V staining and caspase 3/7 activation, indicating apoptosis induction. Caspase 3/7 activation and loss of structural integrity were also observed in CRC cell lines grown as spheroids. Drug treatment at clinically relevant concentrations significantly reduced the viability of patient-derived CRC organoids. Given these in vitro findings, perhexiline, as a racemic mixture or its enantiomers, warrants further investigation as a repurposed drug for use in the management of CRC.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography