Cargando…

Molecularly annotation of mouse avatar models derived from patients with colorectal cancer liver metastasis

Background: Liver is the most common metastatic site in advanced colorectal cancer. Most patients with colorectal cancer liver metastasis (CRLM) do not benefit from current treatment. Patient-derived xenografts (PDXs) with defined molecular signatures are attractive models for preclinical studies. M...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Jingyuan, Xing, Baocai, Liu, Wei, Li, Jian, Wang, Xicheng, Li, Juan, Yang, Jing, Ji, Congcong, Li, Zhongwu, Dong, Bin, Gao, Jing, Shen, Lin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6587174/
https://www.ncbi.nlm.nih.gov/pubmed/31281492
http://dx.doi.org/10.7150/thno.32033
_version_ 1783429018023886848
author Wang, Jingyuan
Xing, Baocai
Liu, Wei
Li, Jian
Wang, Xicheng
Li, Juan
Yang, Jing
Ji, Congcong
Li, Zhongwu
Dong, Bin
Gao, Jing
Shen, Lin
author_facet Wang, Jingyuan
Xing, Baocai
Liu, Wei
Li, Jian
Wang, Xicheng
Li, Juan
Yang, Jing
Ji, Congcong
Li, Zhongwu
Dong, Bin
Gao, Jing
Shen, Lin
author_sort Wang, Jingyuan
collection PubMed
description Background: Liver is the most common metastatic site in advanced colorectal cancer. Most patients with colorectal cancer liver metastasis (CRLM) do not benefit from current treatment. Patient-derived xenografts (PDXs) with defined molecular signatures are attractive models for preclinical studies. Methods: Successfully established PDXs were evaluated to elucidate their fidelity of patients' biologic characteristics (pathologic, genetic and protein properties, together with chemosensitivity). The genomic variations of PDXs were analyzed by next-generation sequencing to explore the underlying molecular mechanism of metastasis and potential therapeutic targets. Results: CRLM (N=73) showed a significantly higher successful PDX establishment rate than primary specimens (N=26; 76.7% vs. 57.7%). CRLM PDXs recapitulated the pathologic, genetic and protein properties of parental tumors, as well as chemosensitivity. Frequent altered genes in PDXs showed high consistency compared to patients' genomic alterations and were enriched in MAPK, ErbB, cell cycle, focal adhesion pathways for CRLM PDXs, whereas primary tumor-derived PDXs only exhibited genomic variations involving ErbB and cell cycle. The genetic alterations showed high concordance between paired PDXs from primary and metastatic tissues, except for recurrent gene mutations (ARID1A, CDK8, ETV1, STAT5B and WNK3) and common copy number gains in chromosomes 20q (e.g., SRC/AURKA). Several potential drug targets such as KRAS, HER2, and FGFR2 were validated using corresponding inhibitors. Additionally, PDX models could also be used in screening efficient regimens for patients with no druggable alterations. Conclusion: This study has successfully established and validated a large panel of molecularly annotated platforms from patients with CRLM for preclinical studies.
format Online
Article
Text
id pubmed-6587174
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-65871742019-07-05 Molecularly annotation of mouse avatar models derived from patients with colorectal cancer liver metastasis Wang, Jingyuan Xing, Baocai Liu, Wei Li, Jian Wang, Xicheng Li, Juan Yang, Jing Ji, Congcong Li, Zhongwu Dong, Bin Gao, Jing Shen, Lin Theranostics Research Paper Background: Liver is the most common metastatic site in advanced colorectal cancer. Most patients with colorectal cancer liver metastasis (CRLM) do not benefit from current treatment. Patient-derived xenografts (PDXs) with defined molecular signatures are attractive models for preclinical studies. Methods: Successfully established PDXs were evaluated to elucidate their fidelity of patients' biologic characteristics (pathologic, genetic and protein properties, together with chemosensitivity). The genomic variations of PDXs were analyzed by next-generation sequencing to explore the underlying molecular mechanism of metastasis and potential therapeutic targets. Results: CRLM (N=73) showed a significantly higher successful PDX establishment rate than primary specimens (N=26; 76.7% vs. 57.7%). CRLM PDXs recapitulated the pathologic, genetic and protein properties of parental tumors, as well as chemosensitivity. Frequent altered genes in PDXs showed high consistency compared to patients' genomic alterations and were enriched in MAPK, ErbB, cell cycle, focal adhesion pathways for CRLM PDXs, whereas primary tumor-derived PDXs only exhibited genomic variations involving ErbB and cell cycle. The genetic alterations showed high concordance between paired PDXs from primary and metastatic tissues, except for recurrent gene mutations (ARID1A, CDK8, ETV1, STAT5B and WNK3) and common copy number gains in chromosomes 20q (e.g., SRC/AURKA). Several potential drug targets such as KRAS, HER2, and FGFR2 were validated using corresponding inhibitors. Additionally, PDX models could also be used in screening efficient regimens for patients with no druggable alterations. Conclusion: This study has successfully established and validated a large panel of molecularly annotated platforms from patients with CRLM for preclinical studies. Ivyspring International Publisher 2019-05-25 /pmc/articles/PMC6587174/ /pubmed/31281492 http://dx.doi.org/10.7150/thno.32033 Text en © Ivyspring International Publisher This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Wang, Jingyuan
Xing, Baocai
Liu, Wei
Li, Jian
Wang, Xicheng
Li, Juan
Yang, Jing
Ji, Congcong
Li, Zhongwu
Dong, Bin
Gao, Jing
Shen, Lin
Molecularly annotation of mouse avatar models derived from patients with colorectal cancer liver metastasis
title Molecularly annotation of mouse avatar models derived from patients with colorectal cancer liver metastasis
title_full Molecularly annotation of mouse avatar models derived from patients with colorectal cancer liver metastasis
title_fullStr Molecularly annotation of mouse avatar models derived from patients with colorectal cancer liver metastasis
title_full_unstemmed Molecularly annotation of mouse avatar models derived from patients with colorectal cancer liver metastasis
title_short Molecularly annotation of mouse avatar models derived from patients with colorectal cancer liver metastasis
title_sort molecularly annotation of mouse avatar models derived from patients with colorectal cancer liver metastasis
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6587174/
https://www.ncbi.nlm.nih.gov/pubmed/31281492
http://dx.doi.org/10.7150/thno.32033
work_keys_str_mv AT wangjingyuan molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT xingbaocai molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT liuwei molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT lijian molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT wangxicheng molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT lijuan molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT yangjing molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT jicongcong molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT lizhongwu molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT dongbin molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT gaojing molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis
AT shenlin molecularlyannotationofmouseavatarmodelsderivedfrompatientswithcolorectalcancerlivermetastasis