[HTML][HTML] Phenotypic and transcriptional fidelity of patient-derived colon cancer xenografts in immune-deficient mice

J Chou, MP Fitzgibbon, CLL Mortales, AMH Towlerton… - PloS one, 2013 - journals.plos.org
J Chou, MP Fitzgibbon, CLL Mortales, AMH Towlerton, MP Upton, RS Yeung, MW McIntosh…
PloS one, 2013journals.plos.org
Xenografts of human colorectal cancer (CRC) in immune-deficient mice have great potential
for accelerating the study of tumor biology and therapy. We evaluated xenografts
established in NOD/scid/IL2Rγ-null mice from the primary or metastatic tumors of 27 patients
with CRC to estimate their capacity for expanding tumor cells for in vitro studies and to
assess how faithfully they recapitulated the transcriptional profile of their parental tumors.
RNA-seq analysis of parental human CRC tumors and their derivative xenografts …
Xenografts of human colorectal cancer (CRC) in immune-deficient mice have great potential for accelerating the study of tumor biology and therapy. We evaluated xenografts established in NOD/scid/IL2Rγ-null mice from the primary or metastatic tumors of 27 patients with CRC to estimate their capacity for expanding tumor cells for in vitro studies and to assess how faithfully they recapitulated the transcriptional profile of their parental tumors. RNA-seq analysis of parental human CRC tumors and their derivative xenografts demonstrated that reproducible transcriptional changes characterize the human tumor to murine xenograft transition. In most but not all cases, the human stroma, vasculature, and hematopoietic elements were systematically replaced by murine analogues while the carcinoma component persisted. Once established as xenografts, human CRC cells that could be propagated by serial transplantation remained transcriptionally stable. Three histologically atypical xenografts, established from patients with peritoneal metastases, contained abundant human stromal elements and blood vessels in addition to human tumor cells. The transcriptomes of these mixed tumor/stromal xenografts did not closely resemble those of their parental tumors, and attempts to propagate such xenografts by serial transplantation were unsuccessful. Stable expression of numerous genes previously identified as high priority targets for immunotherapy was observed in most xenograft lineages. Aberrant expression in CRC cells of human genes that are normally only expressed in hematopoietic cells was also observed. Our results suggest that human CRC cells expanded in murine xenografts have great utility for studies of tumor immunobiology and targeted therapies such as immunotherapy but also identify potential limitations.
PLOS