Activating BRAF and PIK3CA Mutations Cooperate ...

4 downloads 0 Views 2MB Size Report
Landman, Eric Collisson, Joseph Juan, and J. Edward van Veen for advice, guidance, .... Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J,.
Published OnlineFirst April 25, 2014; DOI: 10.1158/1541-7786.MCR-14-0158-T

Molecular Cancer Research

Cell Cycle and Senescence

Activating BRAF and PIK3CA Mutations Cooperate to Promote Anaplastic Thyroid Carcinogenesis Roch-Philippe Charles1,2, Jillian Silva1,2, Gioia Iezza3, Wayne A. Phillips4, and Martin McMahon1,2

Abstract Thyroid malignancies are the most common type of endocrine tumors. Of the various histologic subtypes, anaplastic thyroid carcinoma (ATC) represents a subset of all cases but is responsible for a significant proportion of thyroid cancer-related mortality. Indeed, ATC is regarded as one of the more aggressive and hard to treat forms of cancer. To date, there is a paucity of relevant model systems to critically evaluate how the signature genetic abnormalities detected in human ATC contribute to disease pathogenesis. Mutational activation of the BRAF protooncogene is detected in approximately 40% of papillary thyroid carcinoma (PTC) and in 25% of ATC. Moreover, in ATC, mutated BRAF is frequently found in combination with gain-of-function mutations in the p110 catalytic subunit of PI30 -Kinase (PIK3CA) or loss-of-function alterations in either the p53 (TP53) or PTEN tumor suppressors. Using mice with conditional, thyrocyte-specific expression of BRAFV600E, we previously developed a model of PTC. However, as in humans, BRAFV600E-induced mouse PTC is indolent and does not lead to rapid development of end-stage disease. Here, we use mice carrying a conditional allele of PIK3CA to demonstrate that, although mutationally activated PIK3CAH1047R is unable to drive transformation on its own, when combined with BRAFV600E in thyrocytes, this leads to development of lethal ATC in mice. Combined, these data demonstrate that the BRAFV600E cooperates with either PIK3CAH1074R or with silencing of the tumorsuppressor PTEN, to promote development of anaplastic thyroid carcinoma. Implications: This genetically relevant mouse model of ATC will be an invaluable platform for preclinical testing of pathway-targeted therapies for the prevention and treatment of thyroid carcinoma. Mol Cancer Res; 12(7); 979–86. 2014 AACR.

Introduction Thyroid cancers derived from follicular epithelial cells are histologically classified into papillary thyroid carcinoma (PTC), follicular or anaplastic thyroid carcinoma (ATC; ref. 1). Although PTC is indolent and readily managed by surgical resection combined with radioiodine therapy, ATC is highly aggressive with more than 50% of ATC patients dying within 1 year after diagnosis (2). Although metastasis is observed in 10% to 20% of patients with ATC, most patients with ATC succumb to locally invasive, inoperable

Authors' Affiliations: 1Helen Diller Family Comprehensive Cancer Center; Departments of 2Cell and Molecular Pharmacology, and 3Pathology, University of California, San Francisco, California; and 4Surgical Oncology Laboratory, Sir Peter MacCallum Cancer Center, University of Melbourne, Victoria, Australia €r Biochemie Corresponding Authors: Roch-Philippe Charles, Institute fu €hlstrasse 28, CH-3012 Bern, und Molekular Medizin, University of Bern, Bu Switzerland. Phone: þ41-31-631-4344; Fax: þ41-31-631-3737; E-mail: [email protected]; and Martin McMahon, Helen Diller Family Comprehensive Cancer Center Research Building, University of California, 1450 Third Street, MC-0128, Room HD-365, San Francisco, CA 94158, [email protected]. doi: 10.1158/1541-7786.MCR-14-0158-T 2014 American Association for Cancer Research.

disease that is largely refractory to conventional chemotherapy (3). Mutationally activated BRAF (commonly T1799!A in exon 15) encoding BRAFV600E is detected in approximately 40% of PTC and 25% of ATC (4). BRAFV600E is a constitutively active protein kinase that activates the ERK1/2 MAPK pathway (5). The importance of mutated BRAF in thyroid cancer maintenance is suggested by responses of patients with thyroid cancer to vemurafenib, a pharmacologic inhibitor of BRAFV600E (6). Moreover, conditional, thyrocyte-specific expression of BRAFV600E in genetically engineered mouse (GEM) models results in PTC (7). However, as in humans, PTC in this model is indolent and does not routinely result in progressively lethal disease. Human ATC displays multiple cooperating mutational events in tumor suppressors and oncogenes such as TP53 (70%–80%), PTEN (10%–20%), BRAF (25%), H-, N- or KRAS (20%–30%), PIK3CA (15%–25%), and CTNNB1 (60%–65%; ref. 8). Hence, by analogy to other cancer types, it is likely that progression to more aggressive disease is due to cooperative interactions between these various genetic abnormalities. To test this, we generated mice with thyrocyte-specific expression of BRAFV600E in conjunction with expression of mutationally activated PIK3CAH1047R, a constitutively activated form of the p110 catalytic subunit of

www.aacrjournals.org

Downloaded from mcr.aacrjournals.org on October 29, 2015. © 2014 American Association for Cancer Research.

979

Published OnlineFirst April 25, 2014; DOI: 10.1158/1541-7786.MCR-14-0158-T

Charles et al.

PI30 -kinase a (9). Expression of PIK3CAH1047R, which is detected in many cancer types, is predicted to promote elevated PI30 -lipid production leading to activation of AKT protein kinases and other PI30 -lipid effectors in the cell (10). In brief, whereas adult-onset, thyrocyte-specific expression of PIK3CAH1047R had no detectable effect on the thyroid, it cooperated dramatically with BRAFV600E such that mice developed rapidly lethal ATC. Similar observations were also made with thyrocyte-specific expression of BRAFV600E combined with PTEN silencing. Using cultured human thyroid cancer cell lines, we demonstrated that these pathways cooperate to regulate the activity of mTOR and the phosphorylation of 4E-BP1. Hence, we propose that this GEM model of ATC, which recapitulates key features of the human disease, will be useful in understanding thyroid cancer progression and modeling the effects of pathwaytargeted therapy in the preclinical setting. Materials and Methods Mouse breeding and manipulation BRaf CA, Pik3calat-1047R, Ptenlox, and Thyroglobulin:: CreERT2 mice were described previously (7, 11; 9, 12). BRafCA mice have been backcrossed in FVB/N in the laboratory for more than 10 generations; all the others have been obtained in C57BL/6 F129–mixed background and crossed in FVB/N since obtained. All the mice considered here are predominantly FVB/N. Thyrocyte-specific activation of CreERT2 activity was achieved by intraperitoneal injection of 1 mg of tamoxifen dissolved in peanut oil into 4week-old mice. Cell lines The 8505c line was cultured as directed in RPMI complemented with 10% FCS [and validated by short tandem repeat (STR) profiling, performed by Microsynth]. Ocut-2 in DMEM complemented with 10% FCS and nonessential amino acids, STR profiles showed that this cell line was not presenting mouse or human contamination and was of female origin as expected from the literature. The STR profile of Ocut-2 did not present any relevant similarities to any registered cell lines of the American Type Culture Collection. Histology and immunofluorescence of mouse thyroid tissue sections Animal experiments were carried out in accordance with protocols approved by the University of California, San Francisco (San Francisco, CA) Institutional Animal Care and Use Committee (IACUC). Mice were anesthetized by intraperitoneal injection before tissue dissection, and then euthanized by section of the abdominal aorta. Thyroids were removed, rinsed in ice-cold PBS and fixed for 4 hours in ZFix (Anatech). Four- to 5-mm sections of formalin-fixed, paraffin-embedded tissues were stained with hematoxylin and eosin or processed for immunofluorescence. Epitope unmasking was performed by boiling slides for 10 minutes in a buffer comprising 10 mmol/L Tris, 0.5 mmol/L EGTA pH 9.0. Primary antibodies were obtained from the listed

980

Mol Cancer Res; 12(7) July 2014

commercial sources and diluted as follow: anti–TTF-1 (1:200; Santa Cruz Biotechnology), anti-Ki67 (1:300; Abcam), anti-CK19 (1:300, TROMA-III, Hybridoma bank, University of Iowa, Iowa, IA), Vimentin (Cell Signaling Technology; 1-200), and anti–Galectin-3 (Abcam; 1:200). Antigen–antibody complexes were detected using either goat anti-rabbit Alexa-488 (1:500) or goat anti-rat Alexa-488 (1:500; Molecular Probes) with slides counterstained with DAPI to visualize nuclear DNA. Ultrasound imaging Mice were anesthetized using 2%(v/v) isofluorane at which time fur around the neck was removed using Veet depilatory cream. Ultrasound images were collected weekly using the Vevo770 system (VisualSonics). Thyroid size was assessed by counting pixels at the largest diameter of the thyroid and converting pixel count into area (mm2) using scaling software internal to the device in combination with ImageJ (NIH, Bethesda, Maryland, USA). Results Pik3calat-1047R (Pik3caLat hereafter) mice carry a conditional allele that expresses normal PIK3CA before Cremediated recombination after which mutationally activated PIK3CAH1047R is expressed from the normal chromosomal locus at physiologically relevant levels of expression (9). To test the effects of thyrocyte-specific expression of PIK3CAH1047R, we treated compound adult Thyro::CreER; Pik3caLat mice with tamoxifen and monitored prospectively for palpable goiter. Even 1 year after tamoxifen administration, mice with thyrocyte-specific expression of PIK3CAH1047R displayed no obvious thyroid abnormalities. Hence, unlike BRAFV600E, mutationally activated PIK3CAH1047R is insufficient to initiate thyroid hypertrophy or tumorigenesis (7). To test whether PIK3CAH1047R would influence BRAFV600E-induced PTC, we initiated tumorigenesis in adult Thyro::CreER; BRaf CA/þ; Pik3caLat/þ and monitored them prospectively for disease (Fig. 1A). At 6.5 months, 2 of 14 mice became emaciated, displayed labored breathing, and had readily palpable thyroid tumors that were 1 cm3 requiring euthanasia. Over the next 2 months 71% of mice (10/14) presented developed similar end-stage disease requiring euthanasia. The experiment was stopped 12.5 months after tumor initiation, a time at which 85% (12/ 14) of the Thyro::CreER; BRaf CA/þ; Pik3caLat/þ mice had been retired. In contrast, only 37% (4/11) of an independent cohort of initiated Thyro::CreER; BRaf CA/þ mice reached end-stage and required euthanasia (tumors 1 cm3). Kaplan–Meier survival analysis clearly demonstrated the shortened survival of mice bearing BRAFV600E/PIK3CAH1047R versus BRAFV600E-expressing thyroid tumors (Fig. 1A, P < 0.01). To further evaluate BRAFV600E/PIK3CAH1047R cooperation in thyroid tumorigenesis, tumor growth was measured in two additional cohorts of Thyro:: CreER; BRaf CA/þ; Pik3caLat/þ or Thyro::CreER; BRaf CA/þ mice using ultrasound imaging 2 months after initiation (Fig. 1B). Mice bearing BRAFV600E/PIK3CAH1047R thyroid

Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on October 29, 2015. © 2014 American Association for Cancer Research.

Published OnlineFirst April 25, 2014; DOI: 10.1158/1541-7786.MCR-14-0158-T

BRAF and PI3K Cooperate to Carry Thyroid Cancer Progression

Figure 1. Combined thyrocyteV600E specific expression of BRAF H1047R and PIK3CA results in lethal thyroid carcinogenesis. A, Kaplan– Meier survival curves comparing mice survival of tamoxifenCA/þ treated Thyro::CreER; BRaf ; Lat/þ and Thyro::CreER; Pik3ca CA/þ BRaf mice (  , P  0.01; logrank test). B, thyroid tumor burden in tamoxifen-treated Thyro::CreER; CA/þ Lat-HR/þ BRaf ; Pik3ca and CA/þ Thyro::CreER; BRaf mice measured by ultrasonography (  , P  0.01; t test) 2.5 months after tumor induction. C, hematotoxylin and eosin (H&E) staining of histologic section of thyroid from CA/þ control BRaf (WT, wild-type): CA/þ Thyro::CreER; BRaf V600E (BRAF ) and Thyro::CreER; Lat/þ H1047R (PI3KCA ) mice, at Pik3ca 40 (top row) and 200 (bottom row) magnifications. D, H&E staining of histologic section of CA/þ thyroid Thyro::CreER; BRaf ; Lat/þ V600E (BRAF Pik3ca H1047R PI3KCA ) showing an area of PTC (left), tracheal airway invasion (middle), and ATC area (right), at 40 (top row) and 200 (bottom row) magnifications.

tumors displayed an approximately 60% greater tumor burden compared with BRAFV600E alone tumors. To assess the effects of oncogene expression on tumor histology, thyroid specimens were prepared from normal mice or mice with thyrocyte-specific expression of PIK3CAH1047R, BRAFV600E, or combined BRAFV600E/ PIK3CAH1047R (Fig. 1C and D). Both normal- and PIK3CAH1047R-expressing thyroid (Fig. 1C) tissue displayed a characteristic follicular architecture composed of a monolayered cuboidal epithelium of thyrocytes. Indeed, even 12 months after PIK3CAH1047R expression, no alteration in thyroid architecture was observed (data not shown).

www.aacrjournals.org

BRAFV600E expression elicited PTC displaying the characteristic papillary thyroid architecture at 6 to 9 months after initiation as described previously (Fig. 1C, middle; ref. 7). In contrast, when BRAFV600E and PIK3CAH1047R were coexpressed, we observed clear evidence of PTC within 3 to 6 months that extended throughout the entire thyroid (Fig. 1D, left). Moreover, these PTC lesions displayed aggressive characteristics with evidence of invasion into surrounding tissues, most notably the trachea (Fig. 1D, middle). Such invasion resulted in reduced airway diameter that was observed in 70% of the animals represented in the Kaplan–Meier survival curve (Fig. 1A). In addition, the

Mol Cancer Res; 12(7) July 2014

Downloaded from mcr.aacrjournals.org on October 29, 2015. © 2014 American Association for Cancer Research.

981

Published OnlineFirst April 25, 2014; DOI: 10.1158/1541-7786.MCR-14-0158-T

Charles et al.

thyroid tumors in 80% of these animals displayed polygonal giant cells and/or spindle-like cells (Fig. 1D, right), which is a characteristic of human ATC. Such features (local invasion and aberrant tumor cytology) were not observed in PTCs induced by expression of BRAFV600E alone (7). To further characterize BRAFV600E/PIK3CAH1047R collaboration in this model, we generated Thyro::CreER; BRaf CA mice that were either wild-type, heterozygous, or homozygous for the conditional Pik3caLat allele. Of note, 2.5 months after initiation, Thyro::CreER; BRafCA; Pik3caLat/þ mice displayed the expected increase in tumor size compared with mice lacking the conditional Pik3caLat allele. As expected, Thyro::CreER; BRafCA; Pik3caLat/Lat mice also displayed an increase in tumor size (37%) compared with mice lacking the Pik3caLat allele (Fig. 2A). However, although the difference in tumor size between mice heterozygous or homozygous for the Pik3caLat allele was not statistically significant (Fig. 2B), mice homozygous for Pik3caLat/Lat died more rapidly than their heterozygous counterparts (Fig. 2A) most likely due to an earlier onset of the tracheal invasion. Human thyroid malignancies are characterized by alterations in marker expression such as galactin-3 (Gal-3) and CK-19 (13). We, therefore, stained BRAFV600E/PIK3CAH1047R–expressing thyroid tumors for expression of these proteins (Fig. 2C). First, we noted that BRAFV600E/PIK3-

CAH1047R–expressing thyroid tumors displayed areas of PTC and ATC. The regions of PTC were generally positive for Gal-3 and CK-19 as described previously (7). In contrast, regions of ATC retained Gal-3 but lacked CK-19 expression. In addition, ATC-like lesions displayed low/no expression of TTF-1 or E-cadherin, increased Vimentin expression and a higher proliferative index (Ki67). As an alternative strategy to confirm collaboration of BRAFV600E with alterations in the PI3K pathway, we generated Thyro::CreER; BRaf CA; Ptenlox/lox mice to initiate BRAFV600E/PTENnull thyroid tumors. Compared with BRAFV600E alone, these mice displayed increased thyroid tumor burden as soon as 1.5 months after initiation (Fig. 3B). This required complete silencing of PTEN expression because the enhanced tumorigenesis was not noted in Thyro::CreER; BRafCA mice heterozygous for Ptenlox (Fig. 3B). As anticipated, mice with BRAFV600E/PTENnull thyroid tumors developed end-stage disease more rapidly than mice with BRAFV600E expression alone (Fig. 3A, P < 0.01). It was notable that Thyro::CreER; BRaf CA mice heterozygous for Ptenlox tended to reach endpoint more rapidly than mice with BRAFV600E expression alone, presumably because of loss-of-heterozygosity of the remaining PtenWT allele (Fig. 3B). At the histologic level, BRAFV600E/PTENnull thyroid tumors displayed similar characteristics as BRAFV600E/

Figure 2. Thyroid cancer development in mice wild-type, heterozygous or homozygous for Lat Pik3ca . A, Kaplan–Meier survival curves comparing tamoxifen-treated: 1. Thyro:: CA/þ , Thyro::CreER; 2. CreER; BRaf CA/þ Lat/þ ; Pik3ca Thyro::CreER; BRaf CA/þ or; 3. Thyro::CreER; BRaf ; Lat/Lat mice after tumor Pik3ca initiation (  , P  0.01; log-rank test). B, tumor burden measured by ultrasonography ( , P  0.05;   , P  0.01; t test). C, immunofluorescence staining for expression of TTF-1, E-cadherin, Vimentin, Ki67, and Cytokeratin 19 in a histologic section of thyroid CA/þ from Thyro::CreER; BRaf ; Lat-HR/þ Pik3ca focusing on areas of PTC or ATC area as indicated. Nuclear DNA was stained blue with DAPI.

982

Mol Cancer Res; 12(7) July 2014

Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on October 29, 2015. © 2014 American Association for Cancer Research.

Published OnlineFirst April 25, 2014; DOI: 10.1158/1541-7786.MCR-14-0158-T

BRAF and PI3K Cooperate to Carry Thyroid Cancer Progression

V600E

Figure 3. PTEN silencing cooperates with BRAF for anaplastic thyroid cancer progression. A, Kaplan–Meier survival curves comparing survival of CA/þ þ/þ V600 CA/þ lox/þ V600E het CA/þ lox/lox tamoxifen-treated: Thyro::CreER; BRaf ; Pten (BRAF ) Thyro::CreER; BRaf ; Pten (BRAF PTEN ) or Thyro::CreER; BRaf ; Pten V600E null (BRAF PTEN ) mice after tumor initiation (  , P  0.01; log-rank test). B, thyroid tumor burden measured by ultrasonography ( , P  0.05; t test) 1.5 CA/þ þ/þ V600E months after tumor induction. C, H&E staining of histologic section of thyroid tumors observed in Thyro::CreER; BRaf ; Pten (BRAF ) at 40 (top) CA/þ lox/lox V600E ; Pten mice (BRAF and 200 (bottom) magnifications. D, H&E staining of histologic section of thyroid tumors observed in Thyro::CreER; BRaf null PTEN ) displaying areas of PTC (left), tracheal airway invasion (middle), or ATC area (right) at 40 (top row) and 200 (bottom row) magnifications.

PIK3CAH1047R–expressing thyroid tumors. Although BRAFV600E/PTENWT thyroids presented expected PTC features after 9.5 months (Fig. 3C), BRAFV600E/PTENNull thyroid tumors displayed a clear PTC phenotype as early as 3.5 months after initiation (Fig. 3D, left). In addition, we also detected evidence of tracheal invasion (Fig. 3D, middle) and progression to ATC (Fig. 3D, right) of BRAFV600E/PTENnull tumors at time points 3.5 to 8 months after tamoxifen induction. To explore biochemical collaboration between BRAFV600E and PI30 -kinase signaling in more mechanistic detail, we used two human BRAF-mutated ATC cell lines: 8505c and Ocut2 (14). Whereas Ocut2 expresses both BRAFV600E and PIK3CAH1047R, 8505c is BRAFV600E/ TP53null. Cells were treated with inhibitors of either MEK1/2 (PD325901; ref. 15) or class I PI30 -kinases

www.aacrjournals.org

(GDC-0941; ref. 16) either alone or in combination with effects on cell proliferation assessed using Crystal Violet staining (Fig. 4A). Although both PD325901 and GDC0941 displayed antiproliferative effects, in general, MEK1/2 inhibition had a more potent inhibitory effect on cell proliferation than inhibition of class I PI30 -kinases. Moreover, combined blockade of both pathways had a more striking effect on cell proliferation compared with the antiproliferative activity of the single agents. In parallel with the cell proliferation assays, cells were treated with PD325901 or GDC-0941, either alone or in combination, for 4 or 24 hours at which time cell extracts were analyzed by immunoblotting (Fig. 4B). At both time points, pathway blockade was highly selective in that PD325901-inhibited pERK1/2 with no effect on pAKTand GDC-0941–inhibited pAKT with no effect on pERK1/

Mol Cancer Res; 12(7) July 2014

Downloaded from mcr.aacrjournals.org on October 29, 2015. © 2014 American Association for Cancer Research.

983

Published OnlineFirst April 25, 2014; DOI: 10.1158/1541-7786.MCR-14-0158-T

Charles et al.

Figure 4. BRAF and PI30 kinase signaling cooperate for human thyroid cancer cell proliferation and regulation of phospho-rpS6 and 4E-BP1. A, V600E WT /PIK3CA ) and 8505c (BRAF V600E H1047R /PIK3CA ) Ocut2 (BRAF human anaplastic thyroid cancer cells were treated with an inhibitor of MEK1/2 (PD325901, 1 mmol/L) or class I PI30 -kinases (GDC-0941, 2.5 mmol/L) either alone or in combination for 5 days at which time the cells were fixed and stained with Crystal violet. B and C, V600E WT /PIK3CA ) and 8505c (BRAF V600E H1047R Ocut2 (BRAF /PIK3CA ) human ATC cells were treated with an inhibitor of MEK1/2 (PD325901, 1 mmol/L) or class I PI30 -kinases (GDC-0941, 2.5 mmol/L) either alone or in combination for either 4 (B) or 24 (C) hours at which time the cells were lysed and protein expression/phosphorylation assessed by immunoblotting as indicated. V600E

2. Because these pathways are reported to coordinately regulate the machinery that regulates the initiation of protein synthesis, we assessed the effects of PD325901 and GDC0941 on the phosphorylation of ribosomal protein S6 (rpS6) and 4E-BP1, that latter of which directly regulates Capdependent protein translation (17). Phosphorylation of rpS6 and 4E-BP1 seemed more sensitive to PI30 -kinase inhibition at 4 or 24 hours in Ocut2 cells compared with 8505c cells, which is likely a reflection of the fact that Ocut2 cells express PIK3CAH1047R. Moreover, p-RPS6 and p-4E-BP1 were sensitive to MEK1/2 inhibition in both cell types but this was not observed until 24 hours after inhibitor addition. However, in Ocut2 cells, we detected cooperative effects of combined MEK1/2 plus PI30 -kinase inhibition on pRPS6 at 4 and 24 hours after inhibitor addition. This was also true for p-4E-BP1 24 hours after inhibitor addition but not at 4 hours. We also detected evidence of cooperative inhibitory effects on p-RPS6 and p-4E-BP1 in 8505c cells 24 hours after inhibitor addition, but the effects were not as striking as those observed in Ocut2 cells. Overall, these data suggest that BRAFV600E and PI30 kinase signaling cooperate for sustained thyroid cancer cell

984

Mol Cancer Res; 12(7) July 2014

proliferation and that mutational activation of PIK3CA may predict for more potent antiproliferative and signaling effects following inhibition of class I PI30 -kinases. Discussion Anaplastic thyroid carcinoma is a difficult disease to treat because it is generally inoperable and resistant to current regimens of chemotherapy. Although there are provocative hints from small numbers of patients from the phase I vemurafenib clinical trial, it remains to be unequivocally demonstrated that BRAF mutational status is prognostic for therapeutic benefit from BRAFV600E inhibitors. However, the potential clinical benefit of vemurafenib might be influenced by altered signaling through ERBB receptors for EGF ligands as demonstrated by others (18). Furthermore, it was recently demonstrated that blockade of BRAFV600E signaling with a MEK1/2 inhibitor (AZD6244) can restore the expression of the sodium-iodide symporter (NIS), and thereby enhance the efficacy of radioiodine therapy in both GEM models and in patients (19, 20). These data, therefore, suggest that combined pathway-targeted and conventional radiotherapy may be a successful strategy for treating latestage thyroid cancer.

Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on October 29, 2015. © 2014 American Association for Cancer Research.

Published OnlineFirst April 25, 2014; DOI: 10.1158/1541-7786.MCR-14-0158-T

BRAF and PI3K Cooperate to Carry Thyroid Cancer Progression

There are a large number of inhibitors of BRAFV600E and PI30 -kinase signaling being developed for the treatment of a wide range of human cancers. However, it remains unclear whether mutational activation of BRAF, PIK3CA, or silencing of PTEN will serve as strong prognostic biomarkers for thyroid cancer patient responses to pathway-targeted therapy, in part because of effects of these agents on intracellular feedback signaling (18). Indeed, although BRAF mutation is prognostic of response in melanoma, that is not true for patients with colorectal cancer (21, 22). The availability of GEM models of a wide range of thyroid cancer types, including the new model of ATC based on a foundation of mutational activation of BRAF plus either mutation of PIK3CA or silencing of PTEN described here, will allow preclinical modeling of combination pathway-targeted, conventional or immunomodulatory cancer therapy. It was notable that expression of mutationally activated PIK3CAH1047R in thyrocytes was largely without effect in the mouse but that it cooperated strongly with mutationally activated BRAFV600E. This is not without precedent because PIK3CAH1047R expression fails to initiate tumorigenesis in melanocytes and in the lung and pancreatic epithelium (23– 25). In contrast, mutationally activated BRAFV600E is sufficient to initiate tumorigenesis in all of the above target tissues. Hence, we propose that mutationally activated BRAFV600E is a potent initiator of tumorigenesis that is also frequently required for tumor maintenance. In contrast, mutationally activated PIK3CAH1047R is a weak initiator of tumorigenesis but greatly promotes tumorigenesis initiated by other oncogenic events (25). This may explain why, to date, pathway-targeted inhibition of PI30 -kinase signaling has not proven successful in patients, even those whose tumors are silenced for PTEN or mutated for PIK3CA. TRP53 is also frequently mutated in anaplastic thyroid carcinoma (50% of the cases). Indeed, separately, McFadden and colleagues have demonstrated that combined expression of BRAFV600E with silencing of TP53 also elicits anaplastic thyroid carcinoma (26). It has also been showed that the combination of TRP53 and PTEN knockouts could lead to ATC in another mouse model (27). This model has a similar latency to the model described here even if it using an embryonic onset of the cre-recombinase (TPO-Cre). This shows that even if cannot exclude p53 alteration in the BRAFV600E PIK3CAH1047R model, subsequent p53 alteration resulting in ATC progression is very unlikely. The cooperation observed between BRAFV600E and PI30 kinase signaling in GEM models was recapitulated, at least in part, by analysis of bona fide human ATC-derived cell lines. It was clear that combined inhibition of both BRAFV600E and PI30 -kinase signaling had more potent antiproliferative effects than that of single-agent inhibition. Moreover, reg-

ulation of key nodes in protein translation, most notably the phosphorylation of 4E-BP1 was clearly under the dual control of both pathways, suggesting that efficient Capdependent protein synthesis requires inputs from at least two pathways in ATC-derived cell lines. Although there was a suggestion that the PIK3CAH1047R-expressing Ocut2 cells were more sensitive to PI30 -kinase, an extended analysis of a larger panel of thyroid cancer cell lines will be required to confirm if there is a genotype–drug response phenotype in this disease. In conclusion, we describe a new mouse model of anaplastic thyroid carcinoma that recapitulates key features of the genetics and pathobiology of the cognate human disease. We propose that this GEM model will be useful for future preclinical studies and to understand the mechanisms by which these pathways cooperate to promote progression of thyroid cancer from an indolent to a lethal disease Disclosure of Potential Conflicts of Interest M. McMahon has received commercial research grant from Novartis, GlaxoSmith-Kline, and Plexxicon, and is a consultant/advisory board member of Abbvie, Sutro Biopharma, and Igenica Inc. No potential conflicts of interest were disclosed by the other authors.

Authors' Contributions Conception and design: R.-P. Charles, M. McMahon Development of methodology: R.-P. Charles Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): R.-P. Charles, J. Silva, W.A. Phillips Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): R.-P. Charles, J. Silva, G. Iezza, M. McMahon Writing, review, and/or revision of the manuscript: R.-P. Charles, M. McMahon Study supervision: M. McMahon

Acknowledgments The authors thank all of the members of the McMahon laboratory, especially Christy Trejo, Teruyuki Muraguchi, Victoria Marsh, Shon Green, Anny Shai, Allison Landman, Eric Collisson, Joseph Juan, and J. Edward van Veen for advice, guidance, and support throughout this project. The authors thank the support for mouse husbandry from Allen Villanueva. The authors also thank Bill Hyun and Jane Gordon from the UCSF Laboratory for Cell Analysis for assistance with microscopy, Cynthia Cowdrey of the BTRC Tissue Core for processing of formalin-fixed samples and Byron Hann and colleagues of the UCSF Preclinical Therapeutics Core for assistance with ultrasound imaging. The authors thank Dr. Lori Friedman (Genentech) for GDC0941, Prof. J.A. Fagin (MSKCC) for providing the 8505c and Ocut-2 cells and Drs. David McFadden and Tyler Jacks (MIT) for ongoing discussion and communicating results prior to publication.

Grant Support R.-P. Charles was supported by a Swiss National Science Foundation Fellowship, J. Silva was supported by Institutional Research and Academic Career Development Award (IRACDA), W.A. Phillips was supported by project grants from the National Health and Medical Research Council of Australia, and M. McMahon was supported by the National Cancer Institute (CA131261). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Received March 27, 2014; accepted April 14, 2014; published OnlineFirst April 25, 2014.

References 1.

Hundahl S, Fleming I, Fremgen A, Menck H. A National Cancer Data Base report on 53,856 cases of thyroid carcinoma

www.aacrjournals.org

treated in the U.S., 1985-1995 [see comments]. Cancer 1998;83: 2638–48.

Mol Cancer Res; 12(7) July 2014

Downloaded from mcr.aacrjournals.org on October 29, 2015. © 2014 American Association for Cancer Research.

985

Published OnlineFirst April 25, 2014; DOI: 10.1158/1541-7786.MCR-14-0158-T

Charles et al.

2. 3.

4. 5. 6.

7.

8. 9.

10. 11.

12.

13.

14.

15.

16.

986

Hadar T, Mor C, Shvero J, Levy R, Segal K. Anaplastic carcinoma of the thyroid. Eur J Surg Oncol 1993;19:511–6. McIver B, Hay I, Giuffrida D, Dvorak C, Grant C, Thompson GB, et al. Anaplastic thyroid carcinoma: a 50-year experience at a single institution. Surgery 2001;130:1028–34. Xing M. BRAF mutation in thyroid cancer. Endocr Relat Cancer 2005;12:245–62. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature 2002;417:949–54. Kim K, Cabanillas M, Lazar A, Williams M, Sanders D, Ilagan JL, et al. Clinical responses to vemurafenib in patients with metastatic papillary thyroid cancer harboring BRAF(V600E) mutation. Thyroid 2013;23: 1277–83. Charles RP, Iezza G, Amendola E, Dankort D, McMahon M. Mutationally activated BRAFV600E elicits papillary thyroid cancer in the adult mouse. Cancer Res 2011;71:3863–71. Xing M. Genetic alterations in the phosphatidylinositol-3 kinase/Akt pathway in thyroid cancer. Thyroid 2010;20:697–706. Kinross KM, Montgomery KG, Kleinschmidt M, Waring P, Ivetac I, Tikoo A, et al. An activating Pik3ca mutation coupled with Pten loss is sufficient to initiate ovarian tumorigenesis in mice. J Clin Invest 2012;122:553–7. Samuels Y, Velculescu VE. Oncogenic mutations of PIK3CA in human cancers. Cell Cycle 2004;3:1221–4. Dankort D, Filenova E, Collado M, Serrano M, Jones K, McMahon M, et al. A new mouse model to explore the initiation, progression, and therapy of BRAFV600E-induced lung tumors. Genes Dev 2007;21: 379–84. Lesche R, Groszer M, Gao J, Wang Y, Messing A, Sun H, et al. Cre/ loxP-mediated inactivation of the murine Pten tumor suppressor gene. Genesis 2002;32:148–9. Barut F, Onak Kandemir N, Bektas S, Bahadir B, Keser S, Ozdamar SO, et al. Universal markers of thyroid malignancies: galectin-3, HBME-1, and cytokeratin-19. Endocr Pathol 2010;21:80–9. Ito T, Seyama T, Hayashi Y, Hayashi T, Dohi K, Mizuno T, et al. Establishment of 2 human thyroid-carcinoma cell-lines (8305c, 8505c) bearing p53 gene-mutations. Int J Oncol 1994;4:583–6. Barrett SD, Bridges AJ, Dudley DT, Saltiel AR, Fergus JH, Flamme CM, et al. The discovery of the benzhydroxamate MEK inhibitors CI-1040 and PD 0325901. Bioorg Med Chem Lett 2008;18:6501–4. Folkes A, Ahmadi K, Alderton W, Alix S, Baker S, Box G, et al. The identification of 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-piperazin-

Mol Cancer Res; 12(7) July 2014

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

27.

1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer. J Med Chem 2008;51:5522–32. She QB, Halilovic E, Ye Q, Zhen W, Shirasawa S, Sasazuki T, et al. 4EBP1 is a key effector of the oncogenic activation of the AKT and ERK signaling pathways that integrates their function in tumors. Cancer Cell 2010;18:39–51. Montero-Conde C, Ruiz-Llorente S, Dominguez J, Knauf J, Viale A, Sherman EJ, et al. Relief of feedback inhibition of HER3 transcription by RAF and MEK inhibitors attenuates their antitumor effects in BRAF mutant thyroid carcinomas. Cancer Discov 2013;3:520–33. Chakravarty D, Santos E, Ryder M, Knauf J, Liao XH, West BL, et al. Small-molecule MAPK inhibitors restore radioiodine incorporation in mouse thyroid cancers with conditional BRAF activation. J Clin Invest 2011;121:4700–11. Ho A, Grewal R, Leboeuf R, Sherman E, Pfister D, Deandreis D, et al. Selumetinib-enhanced radioiodine uptake in advanced thyroid cancer. N Engl J Med 2013;368:623–32. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011;364:2507–16. Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature 2012;483:100–3. Marsh Durban V, Deuker MM, Bosenberg MW, Phillips W, McMahon M. Differential AKT dependency displayed by mouse models of BRAFV600E-initiated melanoma. J Clin Invest 2013;123:5104–18. Trejo CL, Green S, Marsh V, Collisson EA, Iezza G, Phillips WA, et al. Mutationally activated PIK3CA(H1047R) cooperates with BRAF (V600E) to promote lung cancer progression. Cancer Res 2013;73: 6448–61. Collisson EA, Trejo CL, Silva JM, Gu S, Korkola JE, Heiser LM, et al. A central role for RAF–>MEK–>ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov 2012;2:685–93. McFadden D, Vernon A, Santiago P, Martinez-McFaline R, Bhutkar A, Crowley DM, et al. TP53 constrains progression to anaplastic thyroid carcinoma in a Braf-mutant mouse model of papillary thyroid cancer. Proc Natl Acad Sci U S A 2014;111:E1600–9. Antico Arciuch VG, Russo MA, Dima M, Kang KS, Dasrath F, Liao XH, et al. Thyrocyte-specific inactivation of p53 and Pten results in anaplastic thyroid carcinomas faithfully recapitulating human tumors. Oncotarget 2011;2:1109–26.

Molecular Cancer Research

Downloaded from mcr.aacrjournals.org on October 29, 2015. © 2014 American Association for Cancer Research.

Published OnlineFirst April 25, 2014; DOI: 10.1158/1541-7786.MCR-14-0158-T

Activating BRAF and PIK3CA Mutations Cooperate to Promote Anaplastic Thyroid Carcinogenesis Roch-Philippe Charles, Jillian Silva, Gioia Iezza, et al. Mol Cancer Res 2014;12:979-986. Published OnlineFirst April 25, 2014.

Updated version

Cited articles Citing articles

E-mail alerts Reprints and Subscriptions Permissions

Access the most recent version of this article at: doi:10.1158/1541-7786.MCR-14-0158-T

This article cites 27 articles, 7 of which you can access for free at: http://mcr.aacrjournals.org/content/12/7/979.full.html#ref-list-1 This article has been cited by 3 HighWire-hosted articles. Access the articles at: http://mcr.aacrjournals.org/content/12/7/979.full.html#related-urls

Sign up to receive free email-alerts related to this article or journal. To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at [email protected]. To request permission to re-use all or part of this article, contact the AACR Publications Department at [email protected].

Downloaded from mcr.aacrjournals.org on October 29, 2015. © 2014 American Association for Cancer Research.