<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

PDOs, PDXOs, PDOXs or PDXs? Selecting the Best Model to Progress Your Patient-Centric Research

Selecting the right preclinical model to evaluate a drug candidate’s mechanism of action and efficacy can be challenging given the wide variety of available options and their associated time and cost investments. However, selecting the appropriate model can mean the difference between success and failure for a given drug candidate. 3D in vitro organoids are powerful predictive preclinical models that are ideal for bridging the gap between traditional 2D in vitro cell lines and in vivo studies because they offer a more clinically relevant platform than 2D cell lines prior to transitioning to more costly and time-consuming in vivo validation studies.

Published data have confirmed the “biological equivalency” (>90% correlation) in drug response profiles between matched patient-derived xenograft (PDX) models (i.e., the best preclinical in vivo model for predicting clinical efficacy) and 3D in vitro tumor organoids derived from the same tumor. This means that tumor organoids can serve as patient surrogates in high throughput drug screens and other in vitro applications, which can reduce costs and time associated with using in vivo models, while maintaining clinical relevance. This post highlights the value of using highly clinically relevant patient-relevant tumor organoids for advancing your patient-centric research.

What are Tumor Organoids?

Organoids are 3D in vitro models that can be generated from both healthy and diseased tissues. Scientists at Hubrecht Organoid Technology (HUB) have developed specialized protocols to grow organoids directly from a wide variety of primary human tumors (i.e., tumor organoids) that preserve the cancer stem cell (CSC) compartment. These patient-relevant 3D in vitro cultures faithfully recapitulate the phenotypic, morphologic, and genetic features of their original patient tumor over the long term. Data from peer-reviewed studies (for example, here and here) have established HUB tumor organoids as the only clinically relevant 3D in vitro models with the following features:

  • Genomic and phenotypic stability in long-term culture and after cryopreservation,
  • High clinical predictivity, and
  • Ease of scalability relative to in vivo models.

These features mean that tumor organoids can be passaged over long periods of time, biobanked, and later resuscitated from frozen for follow up studies while retaining the phenotypic and genetic features of the parental tumor over multiple generations.

Tumor organoids are produced using tissue directly from patient tumors (i.e., patient-derived organoids [PDOs]) or patient tumor tissue expanded in mice as PDXs (PDX-derived organoids [PDXOs]), and even patient-derived orthotopic xenografts (i.e., PDOXs). Both PDXOs and PDOXs enable the expansion of PDO collections for population studies by generating new tumor organoid models from existing PDXs.

In 2019, Crown Bioscience entered into a strategic partnership with HUB to become the exclusive provider of HUB technology for preclinical cancer drug development and validation.

Should I Use a PDX or Organoid Model?

PDXs are the optimal preclinical in vivo model for predicting clinical efficacy due to their ability to accurately recapitulate the genomic and phenotypic complexity of human tumors. Because PDXs are derived directly from patient tumor tissue, they closely mirror the genetic integrity relative to the original patient tumor. Additionally, collections of PDX models derived from a specific patient population capture the tumor heterogeneity within that population. However, they can be costly, and time consuming to develop and maintain, making them better suited to later stage studies rather than early-stage large scale screens of candidate compounds.

3D in vitro organoids offer the flexibility of in vitro systems, with the predictive power of patient-derived models. Thus, their application extends to a range of in vitro studies, including:

Since PDXOs are biologically equivalent to PDXs, researchers can readily leverage large collections of well-characterized PDXOs (i.e., biobanks) as surrogates for heterogenous cancer patient populations in high throughput drug screens. This shrinks costs and timelines compared to in vivo studies, while maintaining clinical relevance. Compounds identified in PDXO panel screens allow researchers to make decisions earlier as opposed to waiting for late-stage in vivo PDX model studies, and to progress to more targeted in vivo efficacy studies with higher predictive confidence of the in vitro data.

Advantages and Challenges

As noted earlier, 3D in vitro tumor organoids are more clinically relevant than traditional 2D cell lines, while they can often save researchers time and money when compared to in vivo models. Moreover, since most available PDO/PDXO models are also available as PDXs, researchers have direct access to responsive in vivo matched models that can be used complementarily to allow for an efficient transition from early in vitro studies to late-phase animal-based validation trials.

The advantages of using PDOs is highlighted in a study that leveraged them to uncover an innovative therapeutic clinical candidate. In the study, Herpers et al. (supported by scientists at Crown Bioscience) used PDOs in combination with HCI to functionally evaluate complex drug responses. High throughput screening with tumor organoids identified a bispecific antibody that triggers EGFR degradation specifically in LGR5+ cancer stem cells, with minimal toxicity to healthy LGR5+ colon stem cells. The effect of the antibody was further validated in matched in vivo PDOX and PDX models. This example highlights the potential of functionally screening PDO panels with HCI to efficiently uncover drug candidates, while the subsequent in vivo studies in both PDOX and PDX models provides translational confidence in the PDO screening results.

Researchers should also be aware of some of the common challenges associated with tumor organoids. For instance, they can be developed only for solid tumors of epithelial origin, and they lack a tumor microenvironment component, albeit this limitation can be overcome with tumor organoid co-cultures. Co-cultures with non-autologous human immune cells are the most common and are being used to test the allogeneic T cell response/killing of tumor organoids and potency of immune checkpoint inhibitors.

Summary

Tumor organoids developed using HUB protocols faithfully recapitulate the complexities observed in the parental human cancer tissue, and they show high patient-relevance and translatability. The underlying nature of tumor organoids also makes them robust and reproducible across a wide variety of applications important for drug discovery, including high throughput assays, providing efficiency advantages over in vivo PDX models which are better suited for later stage validation studies.

Leveraging well-characterized tumor organoid biobanks and complementary PDX models, researchers are equipped with an in vitro system that bridges the gap between the convenience of traditional 2D cell lines and the predictive power of in vivo models. This can maximize the chances of selecting drug candidates with high translatability and clinical success.


Related Posts