<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

Applications of Genetically Engineered Organoids and Cell Lines

This post explores applications of genetically engineered organoids and cell lines to expand and accelerate anti-cancer drug discovery and development. We first provide a brief overview of two major methods that are used to develop engineered organoids/cell lines (i.e., lentiviral transduction and PiggyBac), followed by typical workflows for generating these models. Several case studies are presented to showcase their application for target validation and in vitro /in vivo efficacy studies.

Why Use Genetically Engineered Organoids and Cell Lines?

Cell lines and organoids can be readily engineered to stably express transgenes, reporter constructs, and targeted genetic alterations. These modifications greatly expand the application of these models. For instance, tumor organoids engineered to express a bioluminescent probe can be used to establish an orthotopic patient-derived xenograft (PDX), allowing for visualizing the tumor response to treatment in real time and evaluating the response in a relevant tumor environment (TME).

Further, specific genetic mutations known to be underrepresented in patient populations can be introduced, thereby generating highly clinically relevant preclinical models that would otherwise not exist. Additionally, development of panels of organoid models featuring specific mutational profiles can facilitate identifying a target patient population and developing precision medicine approaches.

Genetically engineered in vitro models are increasingly being used to enable the following types of studies relevant to drug discovery and development:

  • Mechanism of action
  • Target discovery/validation
  • Drug resistance
  • Treatment response via reporter lines

Two Methods Used to Generate Genetically Engineered In Vitro Models

A variety of published methods can be used to produce stably expressing engineered models. Below we describe two commonly used methodologies: Lentiviral transduction and PiggyBac. These platforms can be used to develop a wide array of engineered models, including:

  • Standard/inducible over-expression
  • Standard/inducible knockdown
  • Knockout
  • Knock-In

CRISPR/Cas9 gene editing is also being used in both platforms. This versatile and highly efficient technology has opened enormous possibilities for targeted genetic engineering of mammalian organoid and cell line models, specifically for inducing specific genetic alterations.

The following figure depicts the general process for developing engineered organoids.

By targeting single stem cells, transgenic organoids can be derived via clonal expansion and cryopreservation (green). In contrast, differentiated cells cannot form new organoids. From Menche & Farin. Strategies for genetic manipulation of adult stem cell-derived organoids. Experimental & Molecular Medicine volume 53, pages 1483–1494 (2021). Available at https://www.nature.com/articles/s12276-021-00609-8. Used under Creative Commons Attribution 4.0 International License: https://creativecommons.org/licenses/by/4.0/

Lentiviral Transduction

Lentiviral transduction is recognized as a highly efficient method for stable modification of both organoids and cell lines. Lentiviruses do not require cell division for transduction, and they integrate their genome into the infected host cells’ genome. This technology is used across a wide array of applications including for the delivery and stable expression of transgenes, small hairpin RNA (shRNA) constructs, and single-guide RNA (sgRNA) expression cassettes that target sequences via CRISPR/Cas9 gene editing.

Several possible challenges should be taken into consideration prior to using the lentiviral transduction method. The random insertion of the viral genome into the host genome can lead to the unpredictable disruption of endogenous genes (albeit to a lesser degree compared to onco-retroviruses), a variable number of integrations, and generally, lentiviral vectors are limited to DNA sequences up to ~8kb. Beyond this size, viral titers may be reduced substantially.

PiggyBac

This is a non-viral platform that is designed as a two-plasmid system where one contains the expression cassette (the transposon) and the other contains the coding region of the PiggyBac transposase to facilitate the integration of the transposon specifically at ‘TTAA’ tetranucleotide sites dispersed across the genome. The following image illustrates the mechanism of PiggyBac transposition.

Mechanism of piggyBac transposition. (a) In the transfected cell, transposase is expressed from the piggyBac transposase expression vector, and it then recognizes and binds to the specific inverted terminal repeats (ITRs) of the transgene vector plasmid and cuts the DNA sequence of the transgene from the original sites. Then, the transgene DNA sequence integrates into the genomic DNA of the target cell. (b) For excision, re-expression of transposase by transfection of the piggyBac transposase expression vector leads to cutting of the transgene at the ITRs in the genomic DNA, which results in removal of the inserted transgene from the genomic DNA. From Zhang, M., et al. (2017). Gene Delivery and Expression Systems in Induced Pluripotent Stem Cells. In: Sasaki, K., Suzuki, O., Takahashi, N. (eds) Interface Oral Health Science 2016. Springer, Singapore. Available at https://doi.org/10.1007/978-981-10-1560-1_11. Used under Creative Commons Attribution 4.0 International License: https://creativecommons.org/licenses/by/4.0/

PiggyBac was first used to generate engineered mammalian cells in 2005, and it was shown to be capable of achieving stable expression of large inserts (up to 14 kb) without decreases in transposition efficiency. Like lentiviruses, this system is also used across a wide array of applications including to facilitate target gene editing via CRISPR/Cas9. PiggyBac is also suitable for use in cells that are difficult to be transduced.

PiggyBac delivers a fully intact genetic cargo into the host genome, allows for multiple transgenes to be stably delivered and expressed, and the number of integration copies can be regulated by titrating the transposon to transposase ratio. There are several potential challenges with PiggyBac including the possibility of introducing mutations into endogenous genes and controlling the copy number in clones.

Typical Workflow for Generating Engineered Models

Typical timelines for producing genetically engineered organoids and cell lines ranges between 16-32 weeks and 11-15 weeks, respectively. The workflows shown below illustrate the major steps and approximate timing of each step involved in the production of the respective engineered models. The variability in timing is related to several factors, including the model system, DNA modification platform, and design and optimization of parameters. The main steps in each workflow include:

  1. Plasmid Design
  2. Transfection or Transduction
  3. Clone Selection and Validation
  4. Cell Line Delivery

Case Studies

Development of EGFR-Del19-T790M-C797S Resistant Cell Line (Lentiviral Transduction)

PC-9 is a lung adenocarcinoma cell line with a deletion in exon 19 of the EGFR gene. As shown in the schematic below, an engineered PC-9-based cell line was developed to overexpress EGFR-Del19-T790M-C797S, one of the most prevalent EGFR mutants identified in osimertinib (AZD9291)-resistant tumors.

Clones were developed and validated using several assays including genetic sequencing. Four clones were selected for further analysis, two medium and two high expression level clones. As shown in the graph below, overexpression of EGFR-Del19-T790M-C797S conferred cell resistance to osimertinib (AZD9291). Read our previous post discussing the preclinical assessment of osimertinib for NSCLC with EGFR exon 20 insertions.

Inducible Gene Knockdown via shRNA (Lentiviral Transduction)

An inducible gene knockdown model was developed in the U2OS cell line. As shown in the image below, the shRNA (shX) successfully knocked down “Target X” in the presence of the inducer doxorubicin (“Doxy”) [see red boxes] beginning at the first timepoint of 24 hours. Subsequent analyses showed that the knockdown of target X significantly inhibited cell growth compared with the negative control group (shNT).

A20-hROR1 (PiggyBac)

After observing that lentiviral transduction in the A20 cell line was inefficient (i.e., low cell viability in the presence of antibiotic selection), hROR1 was successfully introduced into the A20 cell line using PiggyBac. As shown below, a variety of clones with varying hROR1 expression levels were successfully produced. This valuable model can be used to evaluate hROR1-targeted immuno-therapies, and it is currently undergoing in vivo validation.

STK11 Knockout in MC38 by CRISPR (PiggyBac)

The STK11 gene (LKB1) was successfully knocked out in the murine colorectal carcinoma cell line CT26 using CRISPR gene editing via PiggyBac. In vivo validation studies demonstrated that this engineered cell line had significantly faster tumor growth and PD1 efficacy was abolished in the STK11 knockout CT26 model as compared to the parental cell line.

Furthermore, the combination of trametinib and an anti-PD1 agent produced significant antitumor activity against the CT26-STK11 knockout model as shown via linear mixed model analysis (p=0.035 vs. control group).

Liver Cancer Organoid (PDXO LI6677) Engineered to Express Human CD19 and Luciferase

The LI6677 organoid line was engineered to express the human immune cell target CD19 in combination with the reporter luciferase to allow for quantitative evaluation of the antitumor effects of a CD19 CAR-T immunotherapy. To do this, the engineered organoids were co-cultured with the CD10 CAR-T cells over a period of 48 hours and luciferase activity revealed specific killing of tumor organoids by CD19 CAR-T cells.

Conclusion

Organoids and cell lines expand the number of in vitro models available for drug discovery purposes. The application of genetically engineered organoids and cell lines can benefit a wide array of preclinical studies, and with the appropriate level of upfront planning, these models have the potential to enhance the translatability of preclinical data. Prior to generating an engineered model, it is highly advisable to hold early discussions with experts in engineered in vitro models, including experts with a deep understanding of how to circumvent any potential pitfalls so that your model and downstream studies are positioned for success.

Crown Bioscience has extensive experience with engineered organoids and cell lines, and we currently offer 26 engineered organoid models that cover multiple cancer types and can be applied in multiple types of studies, including in vivo imaging, immunotherapy and mechanism of action studies.


Related Posts