<img height="1" width="1" src="https://www.facebook.com/tr?id=1582471781774081&amp;ev=PageView &amp;noscript=1">
  • Menu
  • crown-logo-symbol-1-400x551

Find it Quickly

Get Started

Select the option that best describes what you are looking for

  • Services
  • Models
  • Scientific Information

Search Here For Services

Click Here to Start Over

Search Here For Models

Click Here to Start Over

Search Here For Scientific Information

Click Here to Start Over

In Vitro

Boost oncology drug discovery with XenoBase®, featuring the largest cell line selection and exclusive 3D organoid models. Benefit from OrganoidXplore™ and OmniScreen™ for rapid, in-depth analysis.

Learn More

In Vivo

Enhance drug development with our validated in vivo models, in vitro/ex vivo assays, and in silico modeling. Tailored solutions to optimize your candidates.

Learn More

Tissue

Experience ISO-certified biobanking quality. Access top biospecimens from a global clinical network, annotated by experts for precise research.

Learn More

Biomarkers and Bioanalysis

Leverage our global labs and 150+ scientists for fast, tailored project execution. Benefit from our expertise, cutting-edge tech, and validated workflows for reliable data outcomes.

Learn More

Data Science and Bioinformatics

Harness your data and discover biomarkers with our top bioinformatics expertise. Maximize data value and gain critical insights to accelerate drug discovery and elevate projects.

Learn More

KRAS

Accelerate innovative cancer treatments with our advanced models and precise drug screening for KRAS mutations, efficiently turning insights into clinical breakthroughs.

Learn More

EGFR

Advance translational pharmacology with our diverse pre-clinical models, robust assays, and data science-driven biomarker analysis, multi-omics, and spatial biology.

Learn More

Drug Resistance

Our suite integrates preclinical solutions, bioanalytical read-outs, and multi-omics to uncover drug resistance markers and expedite discovery with our unique four-step strategy.

Learn More

Patient Tissue

Enhance treatments with our human tumor and mouse models, including xenografts and organoids, for accurate cancer biology representation.

Learn More

Bioinformatics

Apply the most appropriate in silico framework to your pharmacology data or historical datasets to elevate your study design and analysis, and to improve your chances of clinical success.

Learn More

Biomarker Analysis

Integrate advanced statistics into your drug development projects to gain significant biological insight into your therapeutic candidate, with our expert team of bioinformaticians.

Learn More

CRISPR/Cas9

Accelerate your discoveries with our reliable CRISPR solutions. Our global CRISPR licenses cover an integrated drug discovery platform for in vitro and in vivo efficacy studies.

Learn More

Genomics

Rely on our experienced genomics services to deliver high quality, interpretable results using highly sensitive PCR-based, real-time PCR, and NGS technologies and advanced data analytics.

Learn More

In Vitro High Content Imaging

Gain more insights into tumor growth and disease progression by leveraging our 2D and 3D fluorescence optical imaging.

Learn More

Mass Spectrometry-based Proteomics

Next-generation ion mobility mass spectrometry (MS)-based proteomics services available globally to help meet your study needs.

Learn More

Ex Vivo Patient Tissue

Gain better insight into the phenotypic response of your therapeutic candidate in organoids and ex vivo patient tissue.

Learn More

Spatial Multi-Omics Analysis

Certified CRO services with NanoString GeoMx Digital Spatial Profiling.

Learn More

Biomarker Discovery

De-risk your drug development with early identification of candidate biomarkers and utilize our biomarker discovery services to optimize clinical trial design.

Learn More

DMPK Services

Rapidly evaluate your molecule’s pharmaceutical and safety properties with our in vivo drug metabolism and pharmacokinetic (DMPK) services to select the most robust drug formulations.

Learn More

Efficacy Testing

Explore how the novel HuGEMM™ and HuCELL™ platforms can assess the efficacy of your molecule and accelerate your immuno-oncology drug discovery programs.

Learn More

Laboratory Services

Employ cutting-edge multi-omics methods to obtain accurate and comprehensive data for optimal data-based decisions.

Learn More

Pharmacology & Bioanalytical Services

Leverage our suite of structural biology services including, recombinant protein expression and protein crystallography, and target validation services including RNAi.

Learn More

Screens

Find the most appropriate screen to accelerate your drug development: discover in vivo screens with MuScreen™ and in vitro cell line screening with OmniScreen™.

Learn More

Toxicology

Carry out safety pharmacology studies as standalone assessments or embedded within our overall toxicological profiling to assess cardiovascular, metabolic and renal/urinary systems.

Learn More

Preclinical Consulting Services

Learn more about how our consulting services can help to support your journey to the clinic.

Learn More

Our Company

Global CRO in California, USA offering preclinical and translational oncology platforms with high-quality in vivo, in vitro, and ex vivo models.

Learn More

Our Purpose

Learn more about the impact we make through our scientific talent, high-quality standards, and innovation.

Learn More

Our Responsibility

We build a sustainable future by supporting employee growth, fostering leadership, and exceeding customer needs. Our values focus on innovation, social responsibility, and community well-being.

Learn More

Meet Our Leadership Team

We build a sustainable future by fostering leadership, employee growth, and exceeding customer needs with innovation and social responsibility.

Learn More

Scientific Advisory Board

Our Scientific Advisory Board of experts shapes our strategy and ensures top scientific standards in research and development.

Learn More

News & Events

Stay updated with Crown Bioscience's latest news, achievements, and announcements. Check our schedule for upcoming events and plan your visit.

Learn More

Career Opportunities

Join us for a fast-paced career addressing life science needs with innovative technologies. Thrive in a respectful, growth-focused environment.

Learn More

Scientific Publications

Access our latest scientific research and peer-reviewed articles. Discover cutting-edge findings and insights driving innovation and excellence in bioscience.

Learn More

Resources

Discover valuable insights and curated materials to support your R&D efforts. Explore the latest trends, innovations, and expertly curated content in bioscience.

Learn More

Blogs

Explore our blogs for the latest insights, research breakthroughs, and industry trends. Stay educated with expert perspectives and in-depth articles driving innovation in bioscience.

Learn More

  • Platforms
  • Target Solutions
  • Technologies
  • Service Types

How to Use Tumor Organoids for Oncology Drug Screening

tumor organoid workflow of oncology high throughput drug screeningReview how, and why, to use tumor organoids in in vitro oncology drug screening programs.

Why Use Organoids for Oncology Drug Screening?

Before we jump straight into how to use tumor organoids for drug screening, one key question to answer is why use organoids – why not continue with the standard in vitro drug screening platforms I’m using now?

The main answer is clinical relevance. Typical drug discovery relies heavily on immortalized cell lines for high throughput screening (HTS), testing both target engagement and cellular activity. While there are many advantages to this approach, continuous maintenance on plastic results in cell line genetic drift, poor translatability, and reduced disease relevance. This all contributes to the high attrition rate currently seen in oncology drug development.

While more predictive models have been developed using clinical material, such as primary cultures ex vivo or patient-derived xenografts (PDX) in vivo, these haven’t been amenable to HTS. Primary cultures are limited by the amount of tissue available and expansion for follow up studies.

PDX produce abundant material in vivo and provide a highly translational platform for late stage drug development, including through mouse clinical trials (MCT). However, the time and cost of PDX development is prohibitive and these models haven’t back translated effectively for high-throughput in vitro use.

Early stage robust and scalable screening platforms are now critically needed to provide clinically-relevant and translatable data to guide drug development, patient selection, and companion diagnostic (CDx) development as early as possible. Organoids have the potential to fill this gap and provide the in vitro preclinical model needed to overcome current screening challenges.

Tumor Organoids

We’ve written extensively in previous posts on the background and development of organoids and tumor organoids. Briefly, patient-derived organoids are generated in vitro from stem cells or progenitor cells grown in 3D. Organoids have the ability to self-organize and self-renew, resulting in the development of “mini-organs” in a dish. These organoids contain multiple differentiated cell lineages exhibiting remarkable similarities to the in vivo organ of origin.

Pioneering work from Hubrecht Organoid Technology (HUB) has generated robust protocols to develop organoids from adult stem cells from both normal and diseased tissue. HUB tumor organoids recapitulate the genomic, morphological, and pathophysiological characteristics of their parental tumor. This provides highly clinically relevant 3D in vitro tumor models for drug discovery.

Tumor organoids are cryopreserved to generate biobanks of models, as well as expanded following resuscitation without losing the organoid’s original identity. This means organoids are a great tool for understanding molecular mechanisms of cancer as well as the development of novel therapeutics. Tumor organoids are also predictive of patient treatment response, therefore providing in vitro models which are much more relevant than standard 2D in vitro platforms.

Drug Screening Using Tumor Organoids

Building the Largest Tumor Organoid Collection to Represent Patient Diversity

To start using tumor organoids for oncology drug screening, the first step is to develop a large panel of organoids covering a wide spectrum of cancer types, similar to the large panels of cell lines currently available for HTS or PDX collections for mouse clinical trials.

Historically, tumor organoids have been derived directly from patient tumors (patient-derived organoids or PDO). While this is still a highly valid technique producing very relevant models for screening, developing the panel of models needed can be time consuming, due to having to source patient tumor tissue from a range of different cancers.

A new approach is to develop PDX-derived organoids (PDXO), from the large collections of in vivo PDX models that are already available and well annotated. This offers a unique opportunity to expand the repertoire of patient-derived organoid models available, including a wider range of cancer indications with a spectrum of mutational profiles and pharmacological response. This includes poorly responsive or resistant tumors.

PDXO are established from the cancer stem cells found within PDX tumors, in the same way that PDO are derived from patient tumors. The developed PDXO and parental PDX show biological equivalence, based on their shared genomic diversity, tumor heterogeneity, histopathology, and response to drugs. PDXOs also maintain stable genomic, morphological, and pathophysiological identities of the corresponding PDX tumor over many passages.

This therefore highlights PDXO as a unique in vitro platform featuring the gold standard patient relevance of PDX models, the scalability of conventional patient-derived organoids, combined with developmental efficiency from the collections of thousands of PDX already generated. This combination provides an ideal platform for large scale drug screens in patient-relevant models, including combination strategy testing, biomarker identification, and CDx development.

PDXO Screen Workflow

Like other typical HTS screening, PDXO drug screening is an automated, highly reproducible, and robust system, aiming to improve efficiency. The added benefit with PDXO screening is being highly predictive in early stage drug development.

Starting with the master biobank of cryopreserved models, PDXO are expanded into a working biobank, ready for HTS. These PDXO biobanks are mycoplasma tested and STR profiled with sufficient banking, resuscitation, and growth profiling performed to enable repeat use of models. Specific organoids can be selected from an online organoid biobank database. Selected PDXO are then expanded as required and seeded into multi-well plates for drug screening.

Screens are run in a similar way to a 2D screen:

  • In a 384-well format
  • Typically generating IC50 values for 8 compounds
  • With positive and negative controls included on each plate.

Test agents are incubated with the organoids for 5 days with a CellTiter-Glo® viability readout or morphology assessment as endpoints. Model expansion and enrollment into the study to IC50 readout and analysis can be as quick as 5 weeks for multiple agents and multiple tumor models. Endpoint readouts can be extended to other physiological parameters, such as apoptosis or high content imaging.

Key Benefits of Tumor Organoids in Drug Screens

There are many benefits to using organoids in general in oncology drug screens, some of which were discussed above. This includes organoids providing increased patient relevance due to preserving original and phenotypic features as well as tumor 3D architecture, all of which are known to influence drug response.

Tumor organoid drug screens also offer high quality readouts, for example, high signal to noise ratio and low intra-plate variation demonstrating the robustness of the platform.

PDXO screening enables a broader evaluation of compounds and their combinations over a large collection of tumor models. Studies can then move directly to in vivo modeling using matched PDX models.

Differences Between Tumor Organoid and Standard 2D Drug Screens

There are some differences from standard 2D screens that need to be considered when planning an organoid screen.

As discussed above, tumor organoids can be cryopreserved then resuscitated for HTS enrolment. This can result in a lead in time slightly longer than standard in vitro cell lines.

The current 384-format for organoid screening is also not as high throughput as standard cell line HTS. This is partly due to the expansion potential of organoids as the culturing and plating criteria for organoids is different to 2D monolayer cultures. For example, the size, density, and morphology of organoids in 3D culture and passaging steps require slightly more time.

Experience and expertise in organoid culturing is therefore a necessity when designing screening protocols as well as genomic, histopathology, and pharmacology profiling of tumor organoids.

Based on the clinical relevance of tumor organoids, it’s clear that organoid screening data will be unmatched by other in vitro screening tools. The availability of such large scale screens are exciting times in early drug discovery.

Conclusion

A lack of preclinical model systems recapitulating primary patient tumors is a major setback in early stage oncology drug development. The use of in vitro tumor organoids in large-scale drug screens can help overcome this providing a robust, scalable, and predictive 3D system to reinvigorate current development workflows.


Related Posts