<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

Are You Doing Proteomics Correctly for Xenografts?

Proteomic profiling is an important aspect of cancer research and drug development. It provides protein abundance information that complements genomics data from next-generation sequencing (NGS).

In this blog post we explore a recently published study that demonstrates how researchers can achieve better proteomic profiling of human tumor xenografts, by separating human and mouse cells prior to protein extraction and experimental data acquisition via mass spectrometry.

The Value of Large-Scale Proteomic Profiling for Cancer Drug Development

Large-scale proteomic profiling provides protein abundance information that complements other -omics information, such as NGS genomic data. For instance, the National Cancer Institute’s Clinical Proteomic Tumor Analysis Consortium project intends to “accelerate the understanding of the molecular basis of cancer through the application of large-scale proteome and genome analysis.”

Recently, various initiatives have started to systematically characterize the proteomes of human xenograft models, which have long been used to drive preclinical cancer research and drug development. In general, these models are established by implanting either human cancer cell lines (CDXs) or patient tumor material (PDX) into immunodeficient mice; each having its own advantages and disadvantages. For example, CDXs can be developed faster and offer high reproducibility and good convenience for genetic engineering, while PDXs are highly clinically relevant and recapitulate the original tumor’s molecular pathology, histology, genomics, and drug response.

Human tumor xenograft models are unique in that they consist of both human cancer cells and mouse noncancerous cells that not only allow researchers to study the molecular basis of cancer but also facilitate studies relevant to cancer drug development such as drug mechanism and biomarker discovery studies. However, this human-mouse mixture can make proteomic profiling of xenografts challenging when it comes to distinguishing human and mouse proteins. This is because human stroma is rapidly replaced by mouse stroma after engraftment.

proteomics

While some studies separate human and mouse cells before proteomic profiling, the vast majority do not, and instead rely on the “run-then-separate”, which is a notoriously difficult and imperfect approach that relies on computational algorithms to do the species-specific peptide assignments. The major challenge in using computational methods is due to the high overlap in peptides derived from human and mouse proteins; referred to as the “common peptide problem”.

While different computational approaches have been developed for species-specific peptide assignments, including the full-peptide, unique-peptide, and human/mouse-only methods, each has limitations, and any variant or combination of the three is unlikely to achieve good performance due to the common peptide problem.

Now, a new study published in Cancer Research Communications and led by scientists at Crown Bioscience and ShanghaiTech University demonstrates how separating human and mouse cells prior to protein extraction and experimental data acquisition via mass spectrometry (MS) can achieve superior proteomic profiling results when using human tumor xenografts where approximately half of the identified peptides are common.

Below is a high-level summary of this important study.

Study Objectives

The investigators evaluated the performance of three computational methods for characterizing and quantifying human protein expression in the presence of mouse proteins.

  1. Full-peptide approach: Both common and species-unique peptides are searched against a unified human and mouse protein database. Common peptides are assigned to one or both species or discarded.

  2. Unique-peptide approach: Common peptides are discarded to allow unambiguous peptide assignment

  3. Human/mouse-only approach: Peptides are searched against only the human or mouse protein database.

The performance of these methods was tested using two different model systems. The first used peptide mixtures derived from two cell lines: human 293T cells and mouse MC38 cells. The second system used liver PDX models with differing levels of mouse contents. Tandem mass tagging (TMT) quantitative proteomics technology was used because it has high sensitivity, specificity, and the multiplexing capability to accommodate 16 samples. TMT identifies and quantitates proteins in different samples using tandem mass spectrometry.

Key Findings

Human Protein Quantification in Human and Mouse Cell Line Mixtures

Shi et al. generated a series of human-mouse mixed cell samples. Human protein quantification was done by searching against a combined human and mouse reference protein database using all mappable peptides (full-peptide), only unique peptides (unique-peptide), or human-only and mouse-only reference protein databases (human/mouse-only).

The first two methods quantified roughly twice as many human as mouse proteins. This was expected, given that the samples contained either purely or primarily human cells. The human-only method (which ignored mouse content) returned ~1,000 more human proteins compared to the other two methods, although many of these were actually common peptides.

The researchers also evaluated the effect of mouse proteins on human protein quantification. Human differentially expressed proteins (DEPs) were identified between samples with different mouse cell percentages. Mouse proteins negatively affected the results. Many identified human DEPs were not truly differentially expressed between the samples and instead were artifacts due to the influence of mouse proteins.

DEPs also rose rapidly as the mouse cell percentage difference increased. This was due to the high percentage of shared peptides, an issue that is unlikely to be resolved by computational methods. The existence of mouse proteins did not significantly affect the results for the full-peptide and unique-peptide methods, but only if the mouse cell percentage was below ~8%. The human-only approach performed less well compared to these two methods.

Human Protein Quantification in PDX Tumors

Much like these cell line experiments, human protein expression was assessed in five liver PDX tumors by different computational methods. Overall, approximately 3,000–4,000 DEPs were detected between the PDX models for either the original or “demoused” tumor samples (the intra-PDX study).

The high number of DEPs reflects the complex and heterogeneous proteomes in individual PDX models, with a strong positive correlation between the number of DEPs and the percentage of mouse cells in the original tumor sample. The investigators reported about 1,000 DEPs between the original and demoused tumor samples, with only 7.8% mouse cells in a liver PDX tumor sample.

In the inter-PDX study, DEPs were compared between demoused and original tumor samples. The original tumors always had roughly 20–30% of human proteins, with real differential expression between two PDXs that were not detected. Furthermore, approximately 30–40% of DEPs were false positives. The analysis showed that profiling of PDX tumors is very sensitive to mouse stroma when quantifying human protein expression, with many false positives and real DEPs that could not be identified in both intra-PDX and inter-PDX comparative studies.

The results of this analysis were consistent with the human-mouse cell line mixture studies. These outcomes demonstrate the risk of both false positives and negatives and that spurious results can arise from xenograft proteomics data that do not separate human and mouse cells.

Conclusion

Proteomic profiling can provide key insights and an abundance of information that complements other -omics data. Based on the recent publication described in this post, the data convincingly demonstrate that separating human and mouse cells prior to protein extraction and experimental data acquisition via mass spectrometry can achieve superior proteomic profiling results when using human tumor xenografts.

Crown Bioscience provides high-quality and reliable proteomics services. Data-independent acquisition (DIA) has gained increasing popularity in the field of bottom-up proteomics, due to higher reproducibility and improved data completeness. View our poster to learn more - “Systematic Evaluation of Label-Free Protein Quantification Pipelines (DDA vs DIA) in 12 Mouse Syngeneic Models.


Related Posts