<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

Where Do Organoids Fit in Preclinical Cancer Modeling?

Tumor-derived cell lines, organoids, PDX - complementary preclinical models for drug screeningand oncology researchThere’s a wide range of new preclinical cancer models being developed, each with their own strengths and best uses. This post looks at 3D cancer organoids and how they fill the gap between simple 2D in vitro assays and in vivo cell line and patient-derived xenograft models.

Integrating 3D Oncology Models with Traditional Preclinical Resources

The inability of 2D in vitro assays to accurately predict in vivo response is one of the contributing factors to the high attrition rate of cancer therapeutics. Therefore, selecting appropriate preclinical models based on similarity to human biology and disease genotype and phenotype has traditionally relied on animal models.

We have recently discussed the new opportunities that 3D in vitro models, such as tumor spheroids and organoids, can offer. This includes building and applying functional three-dimensional in vitro human tumor translational models for oncology research, immunotherapy studies, and drug screening.

With the variety of oncology cell lines and preclinical models available, it’s now essential to know when to use the right model for the right study role. This includes integrating new models such as 3D cancer organoids into existing preclinical strategies, to fill the gaps between 2D in vitro and conventional in vivo assays.

Cancer Cell Lines for Initial Drug Screening

2D in vitro screening is a mainstay of early preclinical development. Cancer cell lines are usually inexpensive, easy to grow, and readily amenable to high-throughout drug screens of vast libraries of small molecule compounds across large cell line panels.

This can provide information about cell-killing potential, while identifying drug targets and potential biomarkers predicting response. The deep understanding we have of the molecular profiles of a large array of cancer cell lines can facilitate their directed application in drug discovery.

However, it is well known that cell lines adapt to growing on plastic and drift from original disease, losing true disease representation and potential predictive ability. 2D cultures also don’t capture the reality of the 3D tumor biology in situ.

Cell Line Derived Xenografts for Early Stage In Vivo Pharmacology

Transplanting cancer cell lines into immunodeficient mouse models generates conventional, cell line derived xenografts which re-establish a more physiological environment. These models provide a good tool for early stage in vivo drug discovery.

Conventional xenografts carry information learned during in vitro screening forward into the next dimension of drug discovery – in vivo pharmacology. In this case, activity discovered in vitro can be assessed in vivo, in the context of host-determined factors such as ADME and pharmacokinetics.

Xenografts can be run quickly, producing results to compare with a wealth of historic data. This can provide a rapid proof-of-concept as to whether your new agent works in a specific indication, or against a specific overexpressed protein. Typical uses for traditional xenografts include:

  • Initial evaluation of multiple potential candidates or a compound series.

  • To evaluate and rank efficacy of a new agent against a known cell line target.

  • To understand aspects of new drug properties or MOA.

Conventional Xenografts Duplicate Cell Culture Issues

As conventional xenografts are generated from cultured cell lines means they suffer from many of the limitations of 2D cell culture. With xenografts, this includes issues with gene expression and protein function influenced by cell stress combined with clonal selection.

While some publications have reported that cell line xenograft tumors share histological characteristics with their corresponding primary tumors, others have found that these tumors fail to recapitulate primary tumor histology. Moreover, depending on the drug target and/or indication, activity of therapeutic agents on traditional xenograft models may have poor correlation with activity in human patients.

For example, low correlation is observed in studies using pancreatic ductal adenocarcinoma (PDAC) xenografts, probably due to the dense desmoplastic stroma and low blood vessel density found in human PDAC, which are not recapitulated in xenografts of this malignancy.

In this case, genetically engineered mouse models (GEMM) of PDAC offer an alternative model. PDAC GEMM demonstrate similar features to human PDAC, characterized by a dense stroma, which is responsible for a high interstitial pressure and collapsed vessels.

Patient-Derived Xenografts (PDX) For More Predictive In Vivo Assays

PDX models, created by directly implanting patient tumor tissue into immunodeficient mice, removes the issue of in vitro selection pressure. The PDX is then established by serial passage into further animals (P1, Pn, etc.). These models are constantly maintained in vivo, with heterogeneous cell types and a tumor microenvironment closely mimicking that of human tumors.

In this respect, PDX closely recapitulate the genotype and phenotype of patient tumors at establishment. Furthermore, intra-tumor clonal architecture is largely conserved in PDX after serial passaging.

PDX are the ideal tools to use when drug development programs are gearing up for clinical trials, as they have a high degree of translatability - where response to standard of care correlates well with patient clinical response. This provides highly predictive data for guidance on indication or patient clinical stratification and comprehension of resistance mechanisms.

PDX Models Not Instantly Amenable to High Throughput Assays

However, the utilization of the PDX platform is less compatible with large and rapid screening, due to cost and the time needed to establish the PDX tumors in mice. One way to overcome this problem, and to have an integrated drug development program, is to back translate PDX to in vitro and early stage in vivo resources.

Positioned in this respect, cell lines have been derived from PDX maintaining essential histopathological features and genetic profiles of the original patient tumors. This means that any rare fusion or mutation you were targeting in the PDX will still be present for cell screening. This also includes features such as biochemical signaling and response to tumor cell autonomously targeted therapeutics.

The PDX-derived cell lines can also be established as "conventional" xenograft models, again providing the genetic feature of interest in a robust system for early stage drug discovery. PDX-derived cells are also amenable to 3D culture, more closely representing tumor biology in situ. However, establishing these cell lines is also time consuming and is not successful in every case.

Organoids for High Throughput, 3D Large Scale Drug Screening

As we described in more detail in our previous blog, organoids are three-dimensional models of primary tissue obtained from fresh biopsies. Organoids contain stem cells whose self-renewal and differentiation properties make it possible to generate 3D structures containing virtually any cell type.

These cells exhibit spatial organization similar to the corresponding organ and are capable of recapitulating some functions of that organ, providing a highly physiologically relevant system. Moreover, organoids retain cell–cell and cell–matrix interactions that more closely resemble those of the original tumor. Organoids from normal tissues can also be used to characterize any off-target toxicity.

Compared to traditional xenograft models, organoids have higher structural and physiological similarities compared to the patient tumor. This includes a stable genomic profile over extended periods of culture for organoids, and stable drug sensitivity profiles even after prolonged periods in culture.

Compared to PDX models, organoids are established more rapidly and with higher success rate, within a timeframe of 2-3 months between propagation and drug efficacy results. They are accessible for genetic modifications and compatible with large-scale drug screening. Similar drug response patterns are seen for both organoids and PDX models.

PDX models can also be successfully established from tumor organoids, with the matched organoid and PDX showing similar histopathology to the parental tumors from which they were derived.

While PDX models are established and propagated in immunodeficient mice, organoids can also be studied with an immune system component and cocultured with autologous immune cells from the patient to evaluate immunotherapeutics.

Summary

In conclusion, organoids derived from primary tissues fill the gap in the arsenal of preclinical models between 2D cell culture/xenograft and PDX models. As we move forward, organoids are likely to become part of standard preclinical drug development process.


Related Posts