Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Acute Lymphoblastic Leukemia | Research

PDX models reflect the proteome landscape of pediatric acute lymphoblastic leukemia but divert in select pathways

Authors: Anuli C. Uzozie, Enes K. Ergin, Nina Rolf, Janice Tsui, Amanda Lorentzian, Samuel S. H. Weng, Lorenz Nierves, Theodore G. Smith, C. James Lim, Christopher A. Maxwell, Gregor S. D. Reid, Philipp F. Lange

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

Murine xenografts of pediatric leukemia accurately recapitulate genomic aberrations. How this translates to the functional capacity of cells remains unclear. Here, we studied global protein abundance, phosphorylation, and protein maturation by proteolytic processing in 11 pediatric B- and T- cell ALL patients and 19 corresponding xenografts.

Methods

Xenograft models were generated for each pediatric patient leukemia. Mass spectrometry-based methods were used to investigate global protein abundance, protein phosphorylation, and limited proteolysis in paired patient and xenografted pediatric acute B- and T- cell lymphocytic leukemia, as well as in pediatric leukemia cell lines. Targeted next-generation sequencing was utilized to examine genetic abnormalities in patients and in corresponding xenografts. Bioinformatic and statistical analysis were performed to identify functional mechanisms associated with proteins and protein post-translational modifications.

Results

Overall, we found xenograft proteomes to be most equivalent with their patient of origin. Protein level differences that stratified disease subtypes at diagnostic and relapse stages were largely recapitulated in xenografts. As expected, PDXs lacked multiple human leukocyte antigens and complement proteins. We found increased expression of cell cycle proteins indicating a high proliferative capacity of xenografted cells. Structural genomic changes and mutations were reflected at the protein level in patients. In contrast, the post-translational modification landscape was shaped by leukemia type and host and only to a limited degree by the patient of origin. Of 201 known pediatric oncogenic drivers and drug-targetable proteins, the KMT2 protein family showed consistently high variability between patient and corresponding xenografts. Comprehensive N terminomics revealed deregulated proteolytic processing in leukemic cells, in particular from caspase-driven cleavages found in patient cells.

Conclusion

Genomic and host factors shape protein and post-translational modification landscapes differently. This study highlights select areas of diverging biology while confirming murine patient-derived xenografts as a generally accurate model system.
Appendix
Available only for authorised users
Literature
10.
go back to reference Woiterski J, Ebinger M, Witte KE, Goecke B, Heininger V, Philippek M, et al. Engraftment of low numbers of pediatric acute lymphoid and myeloid leukemias into NOD/SCID/IL2Rcγnull mice reflects individual leukemogenecity and highly correlates with clinical outcome. Int J Cancer. 2013;133:1547–56. https://doi.org/10.1002/ijc.28170.CrossRefPubMed Woiterski J, Ebinger M, Witte KE, Goecke B, Heininger V, Philippek M, et al. Engraftment of low numbers of pediatric acute lymphoid and myeloid leukemias into NOD/SCID/IL2Rcγnull mice reflects individual leukemogenecity and highly correlates with clinical outcome. Int J Cancer. 2013;133:1547–56. https://​doi.​org/​10.​1002/​ijc.​28170.CrossRefPubMed
28.
go back to reference Sulong S, Moorman AV, Irving JAE, Strefford JC, Konn ZJ, Case MC, et al. A comprehensive analysis of the CDKN2A gene in childhood acute lymphoblastic leukemia reveals genomic deletion, copy number neutral loss of heterozygosity, and association with specific cytogenetic subgroups. Blood. 2009;113:100–7. https://doi.org/10.1182/blood-2008-07-166801.CrossRefPubMed Sulong S, Moorman AV, Irving JAE, Strefford JC, Konn ZJ, Case MC, et al. A comprehensive analysis of the CDKN2A gene in childhood acute lymphoblastic leukemia reveals genomic deletion, copy number neutral loss of heterozygosity, and association with specific cytogenetic subgroups. Blood. 2009;113:100–7. https://​doi.​org/​10.​1182/​blood-2008-07-166801.CrossRefPubMed
Metadata
Title
PDX models reflect the proteome landscape of pediatric acute lymphoblastic leukemia but divert in select pathways
Authors
Anuli C. Uzozie
Enes K. Ergin
Nina Rolf
Janice Tsui
Amanda Lorentzian
Samuel S. H. Weng
Lorenz Nierves
Theodore G. Smith
C. James Lim
Christopher A. Maxwell
Gregor S. D. Reid
Philipp F. Lange
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-01835-8

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine