Q&A: Applications of Human Intestinal Organoid Cultures in Drug Discovery
Human intestinal organoids provide a scalable and highly predictive model system for studying the effects of novel therapeutics in vitro. Intestinal organoids contain the relevant cell types that constitute the intestinal epithelium and, as such, can be maintained in various states of proliferation or differentiation. These physiologically relevant models reveal the toxicity of compounds that can not be seen in Caco-2 cells, which are frequently used to model the intestine during drug development programs.
In the on-demand webinar, "Applications of Human Intestinal Organoid Cultures in Drug Discovery," Danny Leung and Dr. Victoria Conlin described, in detail, what intestinal organoids are, how they mimic the immunocytochemistry and morphology of the human gut epithelium, and how to use organoids for drug screening. They also discussed the intestinal organoid assays offered by Contract Assay Services (CAS) at º£½ÇÆƽâ°æ Technologies.
Continue reading to find the speaker's answers to questions asked following the live webinar. These answers have been edited for length and clarity.
Meet the Speakers

Dr. Victoria Conlin
Dr. Victoria Conlin obtained her PhD in Respiratory Physiology at Newcastle University, England. Over the last 20 years, she has worked both in academia and industry researching areas of infectious disease and cell biology throughout various epithelial regions of the body. Victoria is passionate about scientific discovery and technological advancements, and her ultimate goal is to develop services that increase the predictive power of how drugs behave in early drug discovery to aid in the increased translation of drugs into humans.

Danny Leung
Danny has 30 years of experience in the field of assay development for pharmacology and toxicology, having worked at various biotechnology companies in the Vancouver area. He holds a BSc in Biological Sciences from Simon Fraser University.
Q&A Report
How do I obtain patient cancer samples from intestinal, lung, and other tissues?
The easiest method is to obtain a biopsy sample at the time of diagnosis. Alternatively, if a tumor is surgically removed, it can also be used as source material for creating organoids. For intestinal tissue, refer to our protocol to isolate human colonic crypts from intestinal biopsies for downstream intestinal organoid culture, as well as the product information sheet for ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ Organoid Growth Medium (Human). To establish a hepatic organoid culture from liver tissue samples, refer to the technical manual for HepatiCultâ„¢ Organoid Kit (Human). If you do not have access to biopsy or tumor samples, biobanks, such as ATCC, NCI, and HUB Organoids, can provide you with relevant patient samples for your research.
Where do you (CAS) get healthy intestinal tissue from?
As part of º£½ÇÆƽâ°æ Technologies’ agreement with HUB Organoids, we have access to the HUB's biobank and can select both healthy and disease-derived tissue from this resource.
How do you measure the cell viability of organoids?
The viability of organoids in 3D Matrigel® cultures can be measured using the CellTiter-Glo® 3D Viability Assay (Promega Catalog #G9683), while monolayer culture viability can be assessed using the CellTiter-Glo® Luminescent Cell Viability Assay (Promega Catalog #G7570). For more information on how to measure cell viability as part of toxicity testing for drug development using intestinal organoids, please refer to our protocol "Toxicity Testing for Drug Development Using Human Intestinal Organoids and ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢".
How do you transform intestinal organoids into a suspension before seeding them as monolayers?
For monolayer studies, dissociating the 3D organoids into single-cell suspensions (as much as possible) at the time of seeding is critical for good monolayer differentiation. We utilize this procedure that combines enzymatic dissociation (0.05% Trypsin ETDA) and mechanical disruption to achieve a cell suspension composed of single cells, or small fragments.
Can you explain why you put organoids in suspension 7 days before applying them to monolayer cultures?
To establish large numbers of organoid monolayers, it is recommended to passage organoids from Matrigel® domes into a suspension culture 7 days prior to establishing monolayer cultures, as sufficient cell numbers are required to seed monolayers. This suspension method allows for the generation of a higher number of organoids in a smaller number of wells to avoid having to combine a large number of separate matrigel domes to seed large numbers of monolayer cultures. Once the organoids are harvested and seeded as monolayers using ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ Monolayer Growth Medium (±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ Organoid Differentiation Medium [Human] + RHO/ROCK Pathway Inhibitor [Y-27632]), the organoids will begin to differentiate and become less proliferative. For more information, refer to section VI of How to Generate Human Intestinal Organoid-Derived Monolayers Using ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ and Passaging Intestinal Organoids into Suspension Culture Using ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢.
How are the different cell types organized when they grow in a monolayer?
When grown in a monolayer, the cells are confluent, polarized, and contain tight junctions. Many diverse cell types are present, including stem cells, enterocytes, and functional goblet cells. In my experience, more proliferative cells and stem cells tend to cluster, as do more differentiated cells. For more information, refer to our research poster on "".
Do we need to culture organoids in a growth or differentiation medium to study intestinal organoid metabolism?
Both are possible, but our internal characterization suggests that cytochrome P450 (CYP450) expression (a key marker for studying metabolism) is higher in organoids cultured in a differentiation medium (±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ Organoid Differentiation Medium [Human]) than those cultured in a growth medium because the CYP activity is found in the enterocytes, which are only present under differentiation conditions. Furthermore, air-liquid interface (ALI)-maintained monolayers, which require differentiation medium, have higher CYP450 activity than those maintained in the submerged state and therefore could be a beneficial model to study intestinal organoid metabolism.
How long can we culture organoids in differentiation media? I need to study Paneth cells for a week, is this possible?
In 3D culture, differentiated cultures can be maintained for up to 2 weeks. In the monolayer format, especially with ALI, cultures can be maintained for up to a few months.
For estimated glomerular filtration rate (eGFR)-targeted compound studies, do you adjust the concentration of growth factors in your medium and how would you recommend your customers approach this?
To make any adjustments to the ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ formulation itself, customers can work with our product and scientific support services to either help plan their experiments to work with our existing products, or to arrange for the manufacture of a custom batch of the medium that is modified to accommodate their experimental plan.
How can you count the organoids? When you seed the organoids, how can you control the uniformity?
Organoid counting is conducted before seeding when the organoids are still in fragments. This process involves using an appropriate cell counting instrument to standardize seeding densities. Accurate seeding is crucial for proper dome and monolayer formation and differentiation. During the dissociation of organoids for seeding, creating a suspension of single cells and small clusters enhances the uniform formation of domes or monolayers. Typically, achieving a confluent monolayer within two days of seeding is necessary to ensure proper differentiation.
How many cell types are found in intestinal organoids?
Once the organoids are differentiated using ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ Organoid Differentiation Medium in either 3D or 2D ALI, we see a mix of intestinal stem cells, transit-amplifying cells, enterocytes, goblet cells, and enteroendocrine cells in roughly physiological proportions. Paneth cells may appear, as well as other rare cell types, such as tuft or microfold cells, but may require external stimuli to promote differentiation and maturation.
Can intestinal organoids reproduce and maintain sex differences?
Yes they can, however, these differences are only those that might be intrinsic to the epithelial cells themselves.
In comparison with cytotoxicity, are barrier integrity effects more or less sensitive? Is that consistent between Caco-2 cells and organoid models?
When using marker molecules to assess barrier integrity, versus a cell viability readout (i.e. CellTiter-Glo®) to assess cytotoxicity, the effect on marker molecule permeability is typically observed at concentrations lower than those that affect viability. This is true for both the Caco-2 and organoid lines, even though the relative sensitivity of Caco-2 and organoids can be quite different and is compound-dependent.
How can we get smaller cell variability percentages and higher z-scores when using organoids for high-throughput screening (HTS)?
Consistent well-to-well variability can be a challenge especially when working with HTS formats and small Matrigel® domes. We have found that achieving a homogeneous suspension of organoid fragments that is periodically resuspended facilitates consistency. Consistent placement and volumes during Matrigel® spotting will also reduce variability (and may be assisted by automation).
Have you calculated the therapeutic index for the positive controls that you tested in your toxicity assays? How do the cell viability half-maximal inhibitory concentrations (IC50s) relate to the therapeutic exposure seen in humans?
The IC50 of control compounds in our assays depends on the type of readout and mechanism of action of the compound in question, as discussed in our webinar, and thus estimations regarding the therapeutic window are subject to the method used to derive them. This may also be compounded by the clinical route of administration of the control compound(s). For example, is the maximum concentration (Cmax) of an IV-administered drug in the blood comparable to the localized gastrointestinal (GI) exposure of an orally delivered drug concerning drug-induced GI toxicity? The data shown in our presentation indicates that the Caco-2 cell line does not indicate toxicity to reference compounds like Colchicine (IC50 > 100 uM), and thus would not be a predictive model for this compound with a known high clinical incidence of diarrhea. The therapeutic index is estimated to be quite large, and ultimately a false negative result for Caco-2.
How do you discriminate the remnant FITC, which is exposed during the experiment, from Lucifer Yellow, the end readout, considering the emission spectrum?
It is true that FITC and Lucifer Yellow have similar emission spectrums and can interfere even though the excitation wavelengths are different. We recommend incorporating a reconditioning/washout step in between the use of each marker, which reduces potential cross-talk.
Have differences in known human pharmacogenomic variants been evaluated in these organoids (response, differences in toxicity, etc.)?
We have used organoids from a variety of donors and we have seen some donor-specific responses. We observed more notable donor-specific differences in colorectal cancer-derived tumor organoids.
Can the organoid analysis software be used for human organoids and does it need a specific microscope? How can we access it?
The organoid imaging analysis algorithm that we use in CAS was developed internally at º£½ÇÆƽâ°æ Technologies and is optimized for mouse organoid morphology. Since human organoids can have a large cystic morphology, individual organoids can appear to overlap when viewed as a 2D image. It is challenging to analyze each organoid object with image-based tools when it is not identifiable as a discrete object. We are in the process of developing new tools and methods to apply image-based analysis to human organoids.
Can you please comment on the availability of this organoid technology? Part of the organoid technology (e.g. supplements in media) seems to be patented. Does this affect your services?
º£½ÇÆƽâ°æ Technologies has a non-exclusive license with HUB Organoids to provide these services to our customers. Customers who use our services do not need any additional licensing to use the data we generate on their behalf. Customers who choose to perform their own experiments in-house using our organoid media products should note these products were developed under a license to intellectual property owned by HUB Organoids. These products are sold for research use only. Purchase of these products does not include the right to use them for drug screening aiming for commercial gain, equipment validation, biobanking, or other commercial purposes. Purchasers wishing to use the HUB Organoid technology for purposes other than basic research use should contact HUB Organoids at to obtain a further license. Purchasers may apply for a license from HUB Organoids, which will not be unreasonably withheld by HUB Organoids.

On-Demand Human Intestinal Organoid Course
Learn the basics of culturing human intestinal organoids and applying them to your research. Our knowledgeable instructors will guide you through the foundations of intestinal organoid culture, including culturing human intestinal organoids, assessing their morphology, understanding their applications, and more.
Are the distances between your three Z-planes when imaging organoids evenly spaced? If so, what is the distance between your three Z-planes?
Our Z-planes are evenly spaced, though it's important to validate appropriate Z-planes that will ensure that every organoid is captured in focus in at least one plane on your instrument. One function of our internally developed algorithm is that the final composited image maintains the focus of the organoid object even if some Z-planes are out of focus, which is critical for accurate object identification.
Have you used gut epithelial cells derived from diseased patients and healthy donors? If so, do you see any differences between these? My question aims to see whether in your organoid culture media you observe the pathogenic phenotype, e.g. inflammatory cytokine secretion, from the "diseased organoid" rather than "healthy organoid".
We have not done these experiments, but publications comparing healthy to inflammatory bowel disease-derived organoids' responses to stimuli suggest that pathogenic phenotypes are maintained in the organoid cultures.
Do you have any data for transporters (P-gp, BCRP) as well as intestinal metabolism (CYP3A4, UGT)?
We have evaluated both P-gp and BCRP transport function in organoid monolayers using substrates and inhibitors, following a protocol based on the —, also summarized in . These efflux studies indicate that organoid-derived monolayers have much greater functional transporter activity compared to Caco-2, though there are also regional differences in activity. For CYP3A4, we have also demonstrated much higher metabolic activity in organoid-derived monolayers compared to Caco-2 monolayers. For UGT and other Phase II metabolic processes, we have not yet characterized those properties in organoid monolayers but that is part of our future planned activities.
What transporters are you seeing on the apical border of your organoids?
We currently have functional transporter data for the efflux transporter substrates methotrexate and quinidine which suggest expression of the BCRP and P-gp transporters, though some non-specificity of the substrates is possible. Confirmation of expression localized to the apical border via immunostaining is in progress.
What in vitro-in vivo (clinical) correlations are established for marker compounds for passive, active transport, and intestinal metabolism?
We are continuously validating our systems using physiologically relevant substrates per FDA guidance and focusing our current efforts on the most clinically relevant mediators of transport and metabolism. If you would like more information on this topic, please reach out to contractassay@stemcell.com.
Is the increase of TEER in ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ Organoid Differentiation Medium (ODM) and differentiated-ALI cultures due to a pseudostratified epithelium rather than monostratified?
Increased differentiation and decreased proliferation is likely the reason for the increase in TransEpithelial Electrical Resistance (TEER) between ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ Organoid Growth Medium vs. ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ Organoid Differentiation Medium. The changes we see in TEER from submerged versus ALI cultures are due to an increase in the secretory cell types present along the crypt-villus axis. The greater the diversity of cell types between secretory and absorptive cells, the weaker the tight junctions, and the more physiologically relevant the system becomes.
Do you have any experience testing permeability with high non-specific binding (NSB) such as PROTACs?
We have not specifically assessed NSB molecules in the barrier integrity assay. Our characterizations in the monolayer model have been limited to molecules with known low passive permeability as a marker to identify loss of barrier integrity, as well as transporter substrates and CYP450 substrates.
What is the best method to culture and stain for high-throughput immunofluorescence assay?
We are currently developing our high-throughput immunofluorescence processes in a monolayer format and that in combination with our burgeoning robotics protocols will vastly increase the scope and scale in which our assays will be run. We don't currently have an organoid immunofluorescence assay that is amenable to high throughput. Our best staining protocols are available as part of our online resources.
How well do these models translate to in vivo animal models and human disease?
The intestinal organoids are comparable to in vivo conditions with their heterogeneous cell populations. Our reference compounds, such as Colchicine and Afatinib, demonstrate toxicity in our organoid assays and these compounds have a high GI toxicity clinical incidence. For more information on the clinical relevance of organoid models, please watch our webinar "Optimizing Organoid Models to Better Predict Clinical Outcomes".
How do you get lower variability and higher consistency in your assays?
We have gone through several optimization processes for our intestinal organoid assays in which we have found ideal seeding densities and technical replicates to maximize consistency and decrease variability. Additionally, using º£½ÇÆƽâ°æ Technologies' high-quality, standardized media products, like our ±õ²Ô³Ù±ð²õ³Ù¾±°ä³Ü±ô³Ùâ„¢ Organoid Growth Medium, significantly contributes to the reliability of our results. Our specialized media are designed to provide uniform performance, reducing variability and ensuring reproducibility across your studies.
Considering the complex and dynamic nature of the human intestinal epithelium, how can we integrate high-content screening techniques with intestinal organoid cultures to enhance the predictability and robustness of drug toxicity and efficacy assessments? Specifically, what are the challenges and potential solutions in quantifying and analyzing the multifaceted responses of organoids to pharmacological agents at a high-throughput scale?
Integrating high-content screening techniques with intestinal organoid assays can significantly improve the predictability and robustness of drug toxicity and efficacy assessments by closely mimicking human intestinal biology. Challenges in this integration include managing the inherent heterogeneity of organoids, standardizing culture conditions, and effectively quantifying complex cellular responses. To mitigate these challenges, using organoid biobanks, such as the HUB, will help reduce variability and manage the inherent heterogeneity of organoids, while our high-quality media products and established protocols ensure our culture conditions are standardized. Additionally, the intestinal organoid viability assay can be adapted to automation to further optimize standardization and we are currently working on the expansion of automation into new capabilities and readout formats.
What characterizations can be done with some of these newer assays that you mentioned towards the end? Are there any potential add-ons?
In addition to the standard readouts, we have begun to characterize cytochrome P450 metabolism and transport function. We have data showing greatly increased CYP450 and transporter activity in organoid-derived monolayers compared to Caco-2 monolayers. Additional characterization, such as gene expression through qPCR and protein expression through immunostaining, may also be performed or requested as an add-on service.
How customizable are CAS assays?
Each study can be customized according to the client's needs. Clients don't have to meet a minimum format with CAS for their services, there's no baseline you need to hit with regards to sample numbers, though of course there can be great economy of scale benefits. Assay parameters, such as the number of test articles, concentrations, replicates, treatment regimen, and duration, are flexible and generally can be accommodated in our workflow.
What is the average number of compounds people submit for CAS assays and how many can be screened?
For study sizes, there is certainly a wide range of requested formats. 6 - 12 compounds tested at 3 - 8 concentrations in triplicate (i.e. 200 - 300 data points) is common, though we can accommodate well over 1,000 data points in a single study with our workflow if needed.
If you have any further questions, contact us via this form or reach out to us at contractassay@stemcell.com.
Explore These Resources
Related Products
Request Pricing
Thank you for your interest in this product. Please provide us with your contact information and your local representative will contact you with a customized quote. Where appropriate, they can also assist you with a(n):
Estimated delivery time for your area
Product sample or exclusive offer
In-lab demonstration