p53 constrains progression to anaplastic thyroid carcinoma in a Braf-mutant mouse model of papillary thyroid cancer.

Titlep53 constrains progression to anaplastic thyroid carcinoma in a Braf-mutant mouse model of papillary thyroid cancer.
Publication TypeJournal Article
Year of Publication2014
AuthorsMcFadden DG, Vernon A, Santiago PM, Martinez-McFaline R, Bhutkar A, Crowley DM, McMahon M, Sadow PM, Jacks T
JournalProc Natl Acad Sci U S A
Volume111
Issue16
PaginationE1600-9
Date Published2014 Apr 22
ISSN1091-6490
KeywordsAnimals, Carcinoma, Carcinoma, Papillary, Cell Differentiation, Cell Proliferation, Disease Models, Animal, Disease Progression, Gene Expression Regulation, Neoplastic, Homozygote, Humans, MAP Kinase Signaling System, Mice, Mice, Transgenic, Mitogen-Activated Protein Kinase Kinases, Mutation, Neoplasm Grading, Protein Kinase Inhibitors, Proto-Oncogene Proteins B-raf, Thyroid Cancer, Papillary, Thyroid Carcinoma, Anaplastic, Thyroid Gland, Thyroid Neoplasms, Thyrotropin, Tumor Suppressor Protein p53
Abstract

Anaplastic thyroid carcinoma (ATC) has among the worst prognoses of any solid malignancy. The low incidence of the disease has in part precluded systematic clinical trials and tissue collection, and there has been little progress in developing effective therapies. v-raf murine sarcoma viral oncogene homolog B (BRAF) and tumor protein p53 (TP53) mutations cooccur in a high proportion of ATCs, particularly those associated with a precursor papillary thyroid carcinoma (PTC). To develop an adult-onset model of BRAF-mutant ATC, we generated a thyroid-specific CreER transgenic mouse. We used a Cre-regulated Braf(V600E) mouse and a conditional Trp53 allelic series to demonstrate that p53 constrains progression from PTC to ATC. Gene expression and immunohistochemical analyses of murine tumors identified the cardinal features of human ATC including loss of differentiation, local invasion, distant metastasis, and rapid lethality. We used small-animal ultrasound imaging to monitor autochthonous tumors and showed that treatment with the selective BRAF inhibitor PLX4720 improved survival but did not lead to tumor regression or suppress signaling through the MAPK pathway. The combination of PLX4720 and the mapk/Erk kinase (MEK) inhibitor PD0325901 more completely suppressed MAPK pathway activation in mouse and human ATC cell lines and improved the structural response and survival of ATC-bearing animals. This model expands the limited repertoire of autochthonous models of clinically aggressive thyroid cancer, and these data suggest that small-molecule MAPK pathway inhibitors hold clinical promise in the treatment of advanced thyroid carcinoma.

DOI10.1073/pnas.1404357111
Alternate JournalProc. Natl. Acad. Sci. U.S.A.
PubMed ID24711431
PubMed Central IDPMC4000830
Grant ListR01 CA131261 / CA / NCI NIH HHS / United States
T32 GM007484 / GM / NIGMS NIH HHS / United States
K08CA160658 / CA / NCI NIH HHS / United States
K08 CA160658 / CA / NCI NIH HHS / United States
R01CA131261 / CA / NCI NIH HHS / United States
/ / Howard Hughes Medical Institute / United States

Person Type: