activationofthemtorpathwayinprimarymedullarythyroid ... · s6, and the eukaryotic translation...

10
Human Cancer Biology Activation of the mTOR Pathway in Primary Medullary Thyroid Carcinoma and Lymph Node Metastases Anna Tamburrino 1 , Alfredo A. Molinolo 3 , Paolo Salerno 1 , Rebecca D. Chernock 4 , Mark Raffeld 2 , Liqiang Xi 2 , J. Silvio Gutkind 3 , Jeffrey F. Moley 4 , Samuel A. Wells, Jr. 1 , and Massimo Santoro 5 Abstract Purpose: Understanding the molecular pathogenesis of medullary thyroid carcinoma (MTC) is prereq- uisite to the design of targeted therapies for patients with advanced disease. Experimental Design: We studied by immunohistochemistry the phosphorylation status of proteins of the RAS/MEK/ERK and PI3K/AKT/mTOR pathways in 53 MTC tissues (18 hereditary, 35 sporadic), including 51 primary MTCs and 2 cases with only lymph node metastases (LNM). We also studied 21 autologous LNMs, matched to 21 primary MTCs. Staining was graded on a 0 to 4 scale (S score) based on the percentage of positive cells. We also studied the functional relevance of the mTOR pathway by measuring cell viability, motility, and tumorigenicity upon mTOR chemical blockade. Results: Phosphorylation of ribosomal protein S6 (pS6), a downstream target of mTOR, was evident (S 1) in 49 (96%) of 51 primary MTC samples. This was associated with activation of AKT (phospho- Ser473, S > 1) in 79% of cases studied. Activation of pS6 was also observed (S 1) in 7 (70%) of 10 hereditary C-cell hyperplasia specimens, possibly representing an early stage of C-cell transformation. It is noteworthy that 22 (96%) of 23 LNMs had a high pS6 positivity (S 3), which was increased compared with autologous matched primary MTCs (P ¼ 0.024). Chemical mTOR blockade blunted viability (P < 0.01), motility (P < 0.01), and tumorigenicity (P < 0.01) of human MTC cells. Conclusion: The AKT/mTOR pathway is activated in MTC, particularly, in LNMs. This pathway sustains malignant features of MTC cell models. These findings suggest that targeting mTOR might be efficacious in patients with advanced MTC. Clin Cancer Res; 18(13); 3532–40. Ó2012 AACR. Introduction Medullary thyroid carcinoma (MTC) arises from neural crest–derived thyroid-C cells and represents 5% to 8% of thyroid cancer cases (1). MTC is sporadic in about 75% of cases and in the remainder it is inherited as a component of the multiple endocrine neoplasia type 2 (MEN2) syn- dromes [MEN2A, MEN2B, and familial MTC (FMTC); ref. 1]. C-cell hyperplasia (CCH), confined to the upper lobes of the thyroid, is a preneoplastic lesion in familial patients with MTC. Some investigators consider CCH an in situ carcinoma (2). In early stages thyroidectomy is curative (3, 4), but unfortunately most patients with hereditary MTC and vir- tually all patients with sporadic MTC present with a clin- ically evident thyroid nodule and are frequently incurable because the cancer has already metastasized to regional lymph nodes or distant sites (1). There had been no effective conventional systemic treatment for metastatic MTC until recently, when clinical trials of tyrosine kinase inhibitors (TKI) showed promising results. The Food and Drug Administration in the United States recently approved the TKI, vandetanib, for the treatment of patients with advanced MTC (5–8). Mutations of the RET ( Rearranged during transfection) gene, which codes for the transmembrane receptor for growth factors of the glial-derived neurotrophic factor (GDNF) family, have been consistently associated with hereditary and sporadic MTC (5). Germline mutations of RET are present in virtually all patients with MEN2 and FMTC, and approximately half of the patients with spo- radic MTC have somatic RET mutations (9, 10). MTC- associated mutations activate the RET kinase and its downstream signaling pathways. Very recently, mutations in RAS, a component of the RET signaling cascade, have been identified in RET-negative sporadic MTC samples (11). Authors' Afliations: 1 Center for Cancer Research, 2 Laboratory of Pathol- ogy, National Cancer Institute; 3 National Institute of Dental and Craniofacial Research, Bethesda, Maryland; 4 Washington University School of Medi- cine, St. Louis, Missouri; and 5 Dipartimento di Biologia e Patologia Cellulare e Molecolare, Universit a di Napoli Federico II, Napoli, Italy Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Samuel A. Wells, Jr., Thyroid Oncology Clinic, Medical Oncology Branch, National Cancer Institute, NIH Building 10, Room 13-240E MSC 1206, 9000 Rockville Pike, Bethesda, MD 20892. Phone: 301-435-7854; Fax: 301-496-9020; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-2700 Ó2012 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 18(13) July 1, 2012 3532 on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Upload: others

Post on 03-Jun-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

Human Cancer Biology

Activationof themTORPathway inPrimaryMedullary ThyroidCarcinoma and Lymph Node Metastases

Anna Tamburrino1, Alfredo A. Molinolo3, Paolo Salerno1, Rebecca D. Chernock4, Mark Raffeld2,Liqiang Xi2, J. Silvio Gutkind3, Jeffrey F. Moley4, Samuel A. Wells, Jr.1, and Massimo Santoro5

AbstractPurpose: Understanding the molecular pathogenesis of medullary thyroid carcinoma (MTC) is prereq-

uisite to the design of targeted therapies for patients with advanced disease.

Experimental Design:We studied by immunohistochemistry the phosphorylation status of proteins of

the RAS/MEK/ERK and PI3K/AKT/mTOR pathways in 53 MTC tissues (18 hereditary, 35 sporadic),

including 51 primary MTCs and 2 cases with only lymph node metastases (LNM). We also studied 21

autologous LNMs,matched to 21 primaryMTCs. Stainingwas graded on a 0 to 4 scale (S score) based on the

percentageof positive cells.Wealso studied the functional relevanceof themTORpathwaybymeasuring cell

viability, motility, and tumorigenicity upon mTOR chemical blockade.

Results: Phosphorylation of ribosomal protein S6 (pS6), a downstream target of mTOR, was evident

(S � 1) in 49 (96%) of 51 primary MTC samples. This was associated with activation of AKT (phospho-

Ser473, S > 1) in 79% of cases studied. Activation of pS6 was also observed (S � 1) in 7 (70%) of 10

hereditary C-cell hyperplasia specimens, possibly representing an early stage of C-cell transformation. It is

noteworthy that 22 (96%)of 23LNMshad ahighpS6positivity (S�3),whichwas increased comparedwith

autologous matched primary MTCs (P ¼ 0.024). Chemical mTOR blockade blunted viability (P < 0.01),

motility (P < 0.01), and tumorigenicity (P < 0.01) of human MTC cells.

Conclusion: The AKT/mTOR pathway is activated in MTC, particularly, in LNMs. This pathway sustains

malignant features of MTC cell models. These findings suggest that targeting mTORmight be efficacious in

patients with advanced MTC. Clin Cancer Res; 18(13); 3532–40. �2012 AACR.

IntroductionMedullary thyroid carcinoma (MTC) arises from neural

crest–derived thyroid-C cells and represents 5% to 8% ofthyroid cancer cases (1). MTC is sporadic in about 75%of cases and in the remainder it is inherited as a componentof the multiple endocrine neoplasia type 2 (MEN2) syn-dromes [MEN2A, MEN2B, and familial MTC (FMTC);ref. 1]. C-cell hyperplasia (CCH), confined to the upperlobes of the thyroid, is a preneoplastic lesion in familialpatients with MTC. Some investigators consider CCH an insitu carcinoma (2).

In early stages thyroidectomy is curative (3, 4), butunfortunately most patients with hereditary MTC and vir-tually all patients with sporadic MTC present with a clin-ically evident thyroid nodule and are frequently incurablebecause the cancer has already metastasized to regionallymphnodes or distant sites (1). There hadbeenno effectiveconventional systemic treatment for metastatic MTC untilrecently, when clinical trials of tyrosine kinase inhibitors(TKI) showed promising results. The Food and DrugAdministration in the United States recently approved theTKI, vandetanib, for the treatment of patientswith advancedMTC (5–8).

Mutations of the RET (Rearranged during transfection)gene, which codes for the transmembrane receptor forgrowth factors of the glial-derived neurotrophic factor(GDNF) family, have been consistently associated withhereditary and sporadic MTC (5). Germline mutations ofRET are present in virtually all patients with MEN2 andFMTC, and approximately half of the patients with spo-radic MTC have somatic RET mutations (9, 10). MTC-associated mutations activate the RET kinase and itsdownstream signaling pathways. Very recently, mutationsin RAS, a component of the RET signaling cascade, havebeen identified in RET-negative sporadic MTC samples(11).

Authors' Affiliations: 1Center for Cancer Research, 2Laboratory of Pathol-ogy,National Cancer Institute; 3National Institute of Dental andCraniofacialResearch, Bethesda, Maryland; 4Washington University School of Medi-cine, St. Louis,Missouri; and 5DipartimentodiBiologia ePatologiaCellularee Molecolare, Universit�a di Napoli Federico II, Napoli, Italy

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

Corresponding Author: Samuel A. Wells, Jr., Thyroid Oncology Clinic,Medical Oncology Branch, National Cancer Institute, NIH Building 10,Room 13-240E MSC 1206, 9000 Rockville Pike, Bethesda, MD 20892.Phone: 301-435-7854; Fax: 301-496-9020; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-11-2700

�2012 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 18(13) July 1, 20123532

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 2: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

RET promotes the activation of several intracellular sig-naling pathways, including the RAS/mitogen-activated pro-tein kinase kinase/extracellular signal–regulated kinase(RAS/MEK/ERK) pathway and the phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway, which in turn promotecell proliferation, invasion, and survival (12–14). PI3Kproduces phosphatidylinositol (3,4,5)-trisphosphate(PIP3) and AKT activation is stimulated through PIP3binding, threonine 308 (Thr308) phosphorylation byPDK1, and, in a positive feed back circuit, through serine473 (Ser473) phosphorylation by the serine/threoninekinase mTOR (12, 15). Inhibition of tuberous sclerosiscomplex 2 (TSC2) by AKT leads to the activation of Rheb,which, in turn, activates mTOR. mTOR triggers cell growthand proliferation by phosphorylating eukaryotic transla-tion regulators, among these p70-S6 kinase (p70S6K orS6K1) that, in turn, phosphorylates the ribosomal proteinS6, and the eukaryotic translation initiation factor 4E–binding protein 1 (4E-BP1; ref. 12). mTOR has receivedconsiderable attention as a therapeutic target. Its pharma-cologic inhibition by rapamycin and its analogues (likeRad001/everolimus) has shown promising preclinical andclinical results in the treatment of many human malignan-cies (16).Here, we studied the phosphorylation status of proteins

of the RAS/MEK/ERK and PI3K/AKT/mTOR pathways insamples of primary MTC, lymph node metastases (LNM),and normal thyroid tissue. The aim of the study was toevaluate activation of these pathways as potential therapeu-tic targets in patients with MTC.

Materials and MethodsHuman MTC samplesArchival, formalin-fixed, paraffin-embedded (FFPE), tis-

sue blocks and slides from cases of primary and pairedmetastatic (when available) MTCs from 1989 to 2009 wereretrieved from the Department of Pathology and Immu-nology at the Washington University School of Medicine(St. Louis, MO). The Institutional Review Boards atWashington University and at the NIH approved the study.Representative hematoxylin and eosin (H&E)-stained sec-tions from each block were evaluated to determine suitabil-

ity for inclusion in the study. The inclusion criteria weretumor size at least 1 to 2 mm in depth and 5 � 5 mm2 inlength and width, appropriate fixation, absence of electro-surgical device lesions, and absence of signs of acidic dec-alcifying agents. By immunohistochemistry analysis all ofthe MTC samples were positive for expression of RET,calcitonin, and carcinoembryonic antigen (CEA; data notshown). The primaryMTCwas available in only 30patients,both the primary MTC and the LNMs were available in 21patients, and LNMs were available in only 2 patients. Forthe majority (37 of 53) of the cases, we also examinednormal thyroid tissue from the thyroid isthmus that isvirtually free of C-cells. In 3 patients, we examined 2independent primary MTC nodules, whereas in 1 patientwe examined 3 independent MTC nodules. A summary ofclinicopathologic features of the cases is shown in Sup-plementary Table S1 (Supplementary Information) More-over, we examined CCH samples in 2 of the hereditaryMTCs and 8 independent cases in which only CCH wasevident histologically (Supplementary Table S2, Supple-mentary Information).

RET and RAS genotypingCores of 0.6 mm in diameter were sampled from FFPE

blocks. Fibrotic or amyloid-rich samples with less than 50%of tumor material were excluded from the analysis. DNAextraction was carried out from deparaffinized FFPE speci-mens using the DNeasy Blood and Tissue Kit (Qiagen).Targeted analysis of RETmutations in exons 10, 11, and 13to 16 was done by pyrosequencing on a PyroMark Q24instrument (Qiagen). Primers for theRET gene pyrosequen-cing reactions were designed in the Laboratory of Pathol-ogy, Center for Cancer Research,NCI,NIHand are availableupon request. Germline RET mutations were present in allof the 18 familial MTC samples and in one CCH sample.The presence of RET mutations in the remaining 7 CCHonly samples was documented previously at the Washing-ton University School of Medicine (data not shown). Tar-geted analysis of RAS mutation was conducted in 28 sam-ples, including 11 hereditary and 17 sporadic patients withMTC (7 RETmutant cases and 10 RETwild-type cases). RASmutations were searched for the 3RAS genes (H-, N-, and K-RAS) by exome sequencing. Briefly, 3 mg of genomic DNAfrom each sample were sheared in 150 to 250 bp fragmentsusing the CovarisTM S220 sonicator (Covaris Inc.). Theends of the DNA fragments were repaired using T4 andKlenow DNA polymerases (NEB) and an A-tail was addedby using Klenow enzyme (NEB). Nucleotide adapters ofabout 100 bp (NEXTflex—BioScientific) specific for eachsample were added at the ends of DNA fragments in areaction buffer containing T4DNA ligase (Enzymatics Inc.).DNA fragments ranging from 250 to 400 bp were purifiedusing the Caliper LabChip XT fractionation system (Cali-per) according to manufacturer’s instructions. The DNAfragments were amplified by PCR using NEXTflex primersaccording to manufacturer’s instructions. PCR productswere purified by using magnetic beads (Agencourt AMPureXP; Beckman) according to manufacturer’s instructions.

Translational RelevanceMedullary thyroid carcinoma (MTC) responds poorly

to conventional chemotherapy or radiation therapy.Understanding the molecular pathways driving MTCformation would be helpful in the design of targetedtherapeutics for patientswith advanced disease.Here,weshow that themTORpathway is consistently activated inMTC, that such activation is robust in MTC metastases,and that the expressionofneoplastic phenotypedependson mTOR activity. Targeting mTOR with rapamycin orits derivatives may be exploited as a therapeutic strategyfor metastatic MTC.

pS6 Phosphorylation in Metastatic MTC

www.aacrjournals.org Clin Cancer Res; 18(13) July 1, 2012 3533

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 3: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

Finally, the quality of the DNA libraries was assayed byusing the 2100 Bioanalyzer Agilent. Equal amounts of eachDNA library were mixed in a hybridization buffer contain-ing biotinylated RNAs complementary to the exons of RASgenes (SureSelect—target enrichment system, Agilent)according to manufacturer’s instructions, and the hybrid-ization reactionwas run for 48hours. Streptavidinmagneticbeads (DynabeadsMyone streptavidinC1; Invitrogen)wereused for DNA/RNA capturing. The captured DNA wasamplified by PCR using NEXTflex primers and quantifiedby real-time PCR. Finally, DNA fragments were sequencedin an Illumina sequencer (Illumina GA II) according tomanufacture’s protocol. The results of the sequences wereanalyzed with the IGV 2.0 software.

Tissue microarraysWe constructed a tissue microarray (TMA) including

tissue samples from 53 patients with MTC (18 hereditaryand 35 sporadic; including 30 primary tumors, 21 pairs ofprimary tumors and LNMs, and 2 independent LNMs)and 37 tissue samples of normal C-cell–free tissue fromthe thyroid isthmus of patients with MTC (Supplemen-tary Table S2, Supplementary Information). From eachFFPE block, one representative core was sampled andmanually embedded in a recipient TMA paraffin blockusing 1 to 2 mm skin biopsy punches. TMA cores featur-ing extensive loss of tissue or damage were excludedfrom the analysis. In the 21 pairs of primary tumorand autologous metastatic lymph nodes, TMA immuno-histochemical data were thereafter confirmed by immu-nohistochemistry for pS6, p-AKT473, and p-ERK on reg-ular 5-mm tissue sections. The 10 CCH samples were allstudied by immunohistochemistry on regular 5-mm tissuesections. Finally, for another 20 primary MTC samplespS6, p-AKT473, and p-ERK immunohistochemical datawere thereafter confirmed and correlated by Spearmantest on regular 5-mm tissue sections.

ImmunohistochemistryThe primary antibodies used in this study are listed in

Supplementary Table S3, Supplementary Information. Allantibodies were tested first in representative MTC samplesby immunohistochemistry on regular 5-mm tissue sec-tions to set optimal staining conditions (antibody dilu-tion and antigen retrieval conditions). Immunohisto-chemical staining of the sections, either cut from theTMA block or from the original FFPE blocks, was carriedout as follows. The tissue slides were immersed in Safe-clear II for dewaxing 3 � 30 minutes, hydrated throughgraded alcohol and distilled water solution, and thenwashed with PBS. Antigen retrieval was carried out byusing 10 mmol/L citric acid in a microwave for 20minutes (2 minutes at 100% power and 18 minutes at10% power). Slides were allowed to cool for 30 minutesat room temperature, rinsed twice with PBS, and incu-bated in 3% hydrogen peroxide for 30 minutes to quenchthe endogenous peroxidase. The sections were thenwashed in distilled water and PBS and incubated in

blocking solution (2.5% bovine serum albumin in PBS)for 1 hour at room temperature. Excess solution wasdiscarded, and the sections were incubated with theprimary antibody diluted in blocking solution at 4�Covernight. After washing with PBS, the slides weresequentially incubated with the biotinylated secondaryantibody (1:400; Vector Laboratories) for 30 minutes,followed by the avidin–biotin complex method (VectorStain Elite, ABC kit; Vector Laboratories) for 30minutes atroom temperature. Finally, slides were washed and devel-oped in 3,30-diaminobenzidine (Sigma FASTDAB tablet;Sigma Chemical) under microscopic control. The reactionwas stopped in tap water, and the tissues were counter-stained with Mayer hematoxylin, dehydrated, andmounted. In all cases, the specificity of the reaction wasvalidated in parallel control sections omitting the primaryantibody. Digital image acquisition was conducted fromeach stained slide by the Aperio ScanScope CS System(Aperio, Inc.). The percentage of stained cells was quan-tified in representative areas by the algorithm Pixel Count.The results were also visually evaluated to check consis-tency of the automated analysis. The results of the quan-tification were converted into a 0 to 4 scale (S score) basedon the percentage of cells which stained positively asfollows: 0, less than 10%; 1, between 10% and 24%; 2,between 25% and 49%; 3, between 50% and 74%; and 4,between 75% and 100%. We conducted a Pearson cor-relation test to exclude that variation in staining intensitywas due to the age of the samples. The test showed nocorrelation between the variables "age of the samples"and "immunohistochemical value" for any of the markersstudied.

Cell culture and reagentsMZ-CRC-1 cells, kindly provided by Dr. Robert F. Gagel

(M.D. AndersonCancer Center, Houston, TX),were derivedfrom a malignant pleural effusion from a patient with ametastatic MTC (17). The cells were originally given to oneof the coauthors (M. Santoro) who provided them to thelead author (A. Tamburrino) in August of 2010. RET cDNAsequencing for MZ-CRC-1 revealed a heterozygous (ATG toACG) transition in RET exon 16 resulting in MEN2B-asso-ciated substitution of threonine 918 for methionine(M918T; ref. 18). The TT cells were obtained fromAmericanType Culture Collection in December 2011. TT cells harbortheMEN2A-associated RET C634Wmutation in exon 11, asconfirmed by genomic DNA sequencing. Each of the celllines is checked periodically (and within the last 6 months)by Western blotting for RET presence and activation and bysequencing for the RET M918T mutation (MZ-CRC-1) andfor the RET C634W mutation (TT). MZ-CRC-1 cells weregrown in Dulbecco’s Modified Eagle’s Medium supplemen-ted with 10% FCS, TT cells were grown in RPMI-1640supplemented with 16% fetal calf serum. Media were sup-plemented with 2 mmol/L L-glutamine and 100 U/mLpenicillin–streptomycin (Gibco, Invitrogen).

Rapamycin was from Calbiochem, everolimus (Rad001)was from LC Laboratories, and Torin 2 was from Tocris

Tamburrino et al.

Clin Cancer Res; 18(13) July 1, 2012 Clinical Cancer Research3534

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 4: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

Bioscience. Vandetanib was provided by AstraZeneca. EGFwas purchased from Sigma.

Cell viability and motility assaysFor viability, MZ-CRC-1 and TT cells were seeded in

quadruplicate in 96-well plates (10,000 cells per well) inculture media with 2.5% and 4% FBS, respectively. After 24hours, cells were treated with the indicated compounds. Atthe indicated time point, cells were incubated for 3 hourswith 10 mL of CellTiter96 AQueous One solution in 100 mLof culture media (Promega) and absorbance was measuredat 490 nm. For migration assays, Transwell inserts withmembranes of 8 mmol/L pore size (Corning-Costar) wereused. Membranes were coated with 10 mg/mL collagen andkept at 4�C overnight. Cells were serum starved for 8 hours,detached by trypsin, and counted by hemocytometer. Atotal of 1 � 105 cells were plated in the upper chambers inserum-freemedium, containing the vehicle or the indicatedcompounds (rapamycin: 100 nmol/L; everolimus/Rad001:100nmol/L, and Torin 2: 100nmol/L). The lower chamberswere filled with MZ-CRC-1 and TT culture media contain-ing 2.5% and 4% FBS, respectively, supplemented with 20ng/mL EGF (Sigma). After 24 hours, cells were fixed inmethanol and stained with hematoxylin. Cells on the topsurface of the membranes were wiped off with cottonswabs. Membranes were removed from the inserts, placedon microscope slides, and images acquired by Scanscope.Migrated cells were counted and the number of migratedcells per mm2 was calculated.

Tumorigenicity assayAnimal studies were carried out according to NIH-

approved protocols in compliance with the NIH Guide forthe Care and Use of Laboratory Animals. A total of 5 � 106

MZ-CRC-1 cells were injected subcutaneously into bothflanksof femalenonobesediabetic/severe combined immu-nodeficient (NOD/SCID) mice (n ¼ 12). When tumorvolume reached approximately 200 mm3 (after 30 daysfrom injection), animals were randomly assigned to receiveintraperitoneal (i.p.) vehicle or rapamycin (5 mg/kg/d).Tumor size was assessed twice a week by caliper and tumorvolumewas calculated according to the formula: (L�W2)/2(where L ¼ length and W ¼ width of tumor). At day 25 oftreatment, 2 hours before sacrifice, mice were given anintraperitoneal injection of bromodeoxyuridine (BrdUrd;BD Pharmingen, 100 mg/kg). Animals were euthanized,and tissue was fixed in 4% paraformaldehyde and embed-ded in paraffin for histologic evaluation and BrdUrdimmunohistochemistry.

Statistical analysisCorrelation among the clinicopathologic parameters and

RET or RAS genotyping was carried out by Fisher exact test.Correlation among the immunohistochemical markers wascarried out by Spearman rank correlation coefficient test.Pearson correlation test was run to assess correlationbetween age of samples and immunohistochemical data.Student t testwas carried out to analyze cell culture andmice

experiments. The software used for all analyses was Prism 5(GraphPad Software, Inc.).

ResultsScreening of the patients’ tumors for RET and RASmutations

The presence of the most common RET mutations inexons 10, 11, and 13 to 16 was evaluated in tumor (orLNMs)DNA in53patientswithMTC(Supplementary TableS2, Supplementary Information; ref. 19). All the 18 MTChereditary cases were positive for RET mutations. Amongthe 35 sporadic cases, 20 were positive for somatic RETmutations (Supplementary Table S2, Supplementary Infor-mation). RAS (N-, H-, and K-RAS)mutations were analyzedin 28 out of the 53 MTCs, including 11 hereditary and 17sporadic (7 RET mutant and 10 RET wild-type) cases. RASmutations (4 HRAS and 2 KRAS) were found in 6 patients,among the 10 sporadic RET-negative samples. RAS and RETmutations were mutually exclusive (Supplementary TableS2, Supplementary Information). Neither the presence northe type of RET and RAS mutation was correlated withgender, age, histologic variant (spindle cells, epithelioid),or other histologic features (fibrosis, amyloid, or necrosis;data not shown).

Ribosomal protein S6 phosphorylation in primaryMTC

We measured by immunohistochemistry the level ofphosphorylation of proteins involved in 2 major signalingcascades: RAS/MEK/ERK, using phospho-ERK Thr202/Tyr204 (p-ERK202/204 antibodies) and PI3K-AKT-mTOR,using phospho-AKT Ser473 or Thr308 (p-AKT473, p-AKT308) and phospho-ribosomal protein S6 Ser235/Ser236(pS6235/236) antibodies. AKT Thr308 is phosphorylated byPDK1, which in turn is stimulated by PI3K, thereforephosphorylation at this site indicates upstream pathwaystimulation. AKT Ser473 is phosphorylated by an mTORcontaining protein complex (TORC2), therefore, phos-phorylation at this site is a marker of mTOR activity(12, 15).

p-ERK202/204, p-AKT473, AKT308 scored negative in allnormal thyroid tissues, whereas pS6 scored weakly positive(S¼ 1) in 7 (21%) of 33 cases of normal thyroid tissue (theremaining 4 normal thyroid tissues were excluded from theanalysis due to extensive tissue loss). In contrast, a signif-icant proportion of MTCs was positive for p-ERK202/204, p-AKT473, andpS6. Indeed, 29 (58%)of 50primaryMTCs and9 (50%) of 18 LNMs stained positively (S > 1) for p-ERK202

(1 primary MTC and 5 LNMs were not evaluable due totissue loss). Notably p-AKT473 staining was mostly nuclear,as shown by the representative images in Fig. 1B. Only 39%of primary MTC samples were p-AKT308-positive (S � 1;Supplementary Table S2, Supplementary Information). Thereduced fraction of p-AKT308–positive cases was likely dueto reduced sensitivity of this antibody; indeed, antibodies top-AKT308 exerted a reduced reactivity compared with anti-bodies to p-AKT473, when AKT phosphorylation was

pS6 Phosphorylation in Metastatic MTC

www.aacrjournals.org Clin Cancer Res; 18(13) July 1, 2012 3535

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 5: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

assessed byWestern blotting in HEK293 cells stimulated byinsulin (data not shown). To confirm the TMA data, wholesections from 20 representative primary MTC samples werestained with pS6, p-ERK, and p-AKT473 antibodies. Spear-man correlation analysis showed a good correlation (P <0.01) between TMA and whole section data for all of thetested antibodies (Supplementary Table S2, SupplementaryInformation).

The upregulation of p-ERK202/204, pS6235/236 andp-AKT473 in MTC compared with normal thyroid wasstatistically significant (P < 0.001). The highest percentageof positive samples was seen with pS6 (Fig. 1A). Indeed,the majority of primary MTCs (49 of 51, 96%) and,notably, all of the LNMs (23 of 23) were pS6-positive(S � 1; Fig. 1A and Supplementary Table S2, Supplemen-tary Information). Representative staining of pS6 from6 MTC samples is shown in Fig. 1B. Overall, pS6 phos-phorylation was homogeneous among different nodulesfrom the same patient, with the exception of one casein which it was strong (S ¼ 4) in one nodule and negative(S ¼ 0) in the other one (data not shown).

A Spearman test was carried out to verify whether therewas a correlation between the different markers. A signif-icant correlation (P¼ 0.006)was found between pS6 and p-AKT473 immunostain; moreover, staining for pS6 and p-AKT473 correlated with p-ERK (P ¼ 0.0002 and P < 0.0001,respectively). The latter finding is consistent with previousreports showing that ERK contributes to mTOR activationvia inhibitory phosphorylation of TSC2, anmTOR-negative

regulator (20). However, some samples (10 of 47) scoredpositive for pS6 but not for p-AKT473. It is possible that inthese samples mTOR activation is dependent on geneticlesions affecting pathways other than PI3K/AKT. There wasno correlation between positivity of p-AKT308 and the othermarkers (P > 0.05).

Finally, no significant difference in pS6, p-AKT, andp-ERK staining was detected between familial/sporadic,RET wild-type/mutant, germline/somatic RET mutantcarriers, M918T/cysteine (609/618/620/630/634) RETmutants, or RAS wild type/mutant samples. However, wecannot exclude that the lack of correlation was due to thereduced sample size. A power calculation showed that tohave a good discrimination (a � 0.05) for pS6 (S > 2) thedifference of the 2 proportions (RET mutant/wild-typesamples) should be at least 0.357, whereas in our series itwas less than 0.1.

Phosphorylated S6 ribosomal protein is detectedin CCH

Seven of 10 CCH samples analyzed were pS6-positive(S � 1). Representative cases are shown in Fig. 2. In 2patients, we examined both the CCH and the primary MTCnodule: thepS6positivitywas increased for both cases in theMTC nodule compared with the CCH. One of these cases(WU20) is shown in Figs. 2 (CCH) and 3 (MTC). Theseresults suggest that activation of the mTOR pathway is anearly event in C-cell transformation and that it furtherincreases along with tumor progression.

Figure 1. ERK and mTOR pathway activation in MTC. A, left, the overall percentage of positive (S � 1) samples for immunohistochemical (IHC) stainingof p-ERK, p-AKT473, and pS6 of primaryMTCs and LNMs. The number of evaluable specimenswas: p-ERK: n¼ 50, p-AKT473: n¼ 47, pS6: n¼ 51 (for primaryMTCs); p-ERK: n ¼ 18, p-AKT473: n ¼ 19, pS6: n ¼ 23 (for LNMs). Right, the S score for each marker. B, representative images of p-AKT473 and pS6immunohistochemistry in primary MTC nodules. Top, IHC for p-AKT473 on samples from patients: WU8 (S ¼ 2), WU13 (S ¼ 2), WU18 (S ¼ 2), WU19 (S ¼ 0),WU20 (S ¼ 2), and WU23 (S ¼ 0). p-AKT473 staining was predominantly nuclear (insets), although many cells displayed membrane or cytoplasmicimmunoreactivity. Bottom, IHC for pS6 on samples from patients WU8 (S¼ 3), WU13 (S¼ 4), WU18 (S¼ 4), WU19 (S¼ 2), WU20 (S¼ 3), and WU23 (S¼ 1).pS6 staining was primarily cytoplasmic (insets). Original magnification: �4; insets: �40.

Tamburrino et al.

Clin Cancer Res; 18(13) July 1, 2012 Clinical Cancer Research3536

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 6: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

Ribosomal protein S6 is prominently phosphorylatedin lymph node metastasesThe positivity for pS6 was high in the LNMs: S score �3

was found in 20 (95%) of 21 LNMs compared with 31(61%) of 51 primary MTCs (for 2 additional patients onlythe LNMs were available; Supplementary Table S2, Supple-mentary Information, Figs. 1B and 3). A Spearman rankcorrelation test showed that the increase in the S score ofLNMs compared with matched primary MTC was statisti-cally significant (r ¼ 0.5, P ¼ 0.024). These results suggestthat activation of the mTOR pathway is particularly strongin MTC metastases.

Inhibition of mTOR decreases viability and motility ofMTC cellsTo assess the effect of mTOR inhibition in vitrowe treated

2 human MTC cell lines, MZ-CRC-1 and TT, with 2 inhi-bitors mainly active on TORC1 (rapamycin and Rad001/everolimus) as well as a double TORC1/TORC2 inhibitor(Torin 2). We could show that in these RET mutant MTC

cells, mTOR is active and its activity depends on RET, beingeffectively inhibited by a RET kinase inhibitor (vandetanib;ref. 18; Supplementary Fig. S1, Supplementary Informa-tion). We measured viability at 3 and 5 days of mTORblockade by MTS assay. After 5 days, the 3 compoundscaused a significant reduction in viability of bothMZ-CRC-1and TT cells (Fig. 4A). We measured cell motility by Trans-wellmotility assay. At 24 hours, Torin 2 exerted a significantreduction of migration of both cell types (P < 0.01); alsorapamycin and Rad001 reduced cell migration, but thiseffect reached statistical significance (P < 0.05) only forMZ-CRC-1 cells (Fig. 4B).

Reduction of MZ-CRC-1 cell tumorigenicity byrapamycin

We injected MZ-CRC-1 cells subcutaneously in bothflanks of NOD/SCID mice (n ¼ 12). When tumors ofapproximately 200 mm3 were detected, animals were ran-domly assigned to receive i.p. rapamycin or vehicle; tumorgrowth was monitored by calipers. After 25 days of

Figure 3. Comparison of pS6staining intensity betweenprimary MTCs and LNMs.Representative images of pS6immunohistochemistry conductedon primary MTC nodules (top) andmatched LNMs (bottom). pS6 scores(S) in the primary tumor and LNMswere, respectively: WU7: 3 and 4,WU34: 2 and 4, WU38: 4and 4, WU41: 2 and 3. Originalmagnification: �4; insets: �40.

Figure 2. pS6 detection in CCH.Top, representative images of pS6staining conducted on full-size CCHslides. Bottom, representativeimages of calcitonin staining (carriedout as a CCH control) of the samefield on an adjacent section. Slideswere from patients: WU17 (pS6: S ¼2), WU20 (pS6: S ¼ 2), WU135 (pS6:S ¼ 2), and WU138 (pS6: S ¼ 2).Original magnification: �4; insets:�40.

pS6 Phosphorylation in Metastatic MTC

www.aacrjournals.org Clin Cancer Res; 18(13) July 1, 2012 3537

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 7: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

treatment, average tumor volume was significantly (P <0.01) reduced by rapamycin treatment compared withcontrol (Fig. 5). In the examined time frame of the exper-iment, metastases were not formed and therefore antimeta-static activity of the treatment could not be tested (notshown).DNA synthesis ratewasmeasured in vivoby BrdUrdincorporation. Rapamycin treatment caused a significantreduction (P < 0.0001) of proliferating cells compared withcontrol.

DiscussionIn this study, we analyzed molecular pathways that are

potentially involved in MTC formation and metastaticdissemination.We found that the AKT/mTOR/pS6 pathwaywas active in the majority of MTC cases. The pathway wasactive early as evidenced by positive staining in familialCCH, suggesting a functional link to the RET mutation,which initiates the familial disease.

Rapa and colleagues recently reported activation of themTOR pathway in MTC (21). The present study confirms

their observation and provides valuable information aboutthe clinical relevance and functional consequences ofmTORpathway activation. Indeed, not only did our presentanalysis reveal that mTOR activation represents an earlyevent in C-cell transformation, but it also shows that AKT/mTOR/pS6 activation was stronger in LNMs than inmatched primary MTCs, suggesting a role of this pathwayin MTC tumor progression in addition to initiation. Fur-thermore, our data show that inhibition of mTOR pathwaycauses significant reduction of MTC cell proliferation,motility, and tumorigenicity. Rad001 effects on TT cellproliferation were consistent with those recently reported(22).

In MTC and CCH, activation of the mTOR/pS6 pathwayis likely mediated by AKT. Indeed, AKT inactivates TSCformed by TSC1 and TSC2 (15). Once associated to TSC1,TSC2 acts as a GTPase-activating protein (GAP) to shutoffthe Rheb1 GTPase. Thus, upon AKT activation, GTP-boundRheb1 promotes mTOR activation (15). In turn, mTORactivates S6K1, that phosphorylates S6, and also feedbacks

Figure4. Effects ofmTORblockadeonMTCcell viability andmotility. A, the bar graphs show the results of cell viability (MTSassay)measurement at days 3 and5 of MZ-CRC-1 and TT cells after treatment with the indicated concentration of rapamycin (Rap), Rad001, or Torin 2 (Tor). The average values oftriplicated experiments are presented with error bars representing the SEM (�, P < 0.05; ��, P < 0.01). B, the bar graphs show the results of cell motility(Transwell assay) measurement at 24 hours of MZ-CRC-1 and TT cells after treatment with the indicated concentration of rapamycin, Rad001, or Torin 2.The average values of triplicated experiments are presented with error bars representing the SEM (�, P < 0.05; ��, P < 0.01).

Tamburrino et al.

Clin Cancer Res; 18(13) July 1, 2012 Clinical Cancer Research3538

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 8: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

positively on AKT by promoting its activation throughSer473 phosphorylation (12, 15). RET is able to stimulateAKT through the recruitment of multiple protein adaptors,such as IRS1/2, FRS2, or SHC-C (23–25), or signalingeffectors, such as SRC (26). Similarly, RAS is also able tostimulate PI3K and therefore AKT (12–14).Wenoted that inMTC, AKT was more consistently phosphorylated onSer473 than on Thr308; while there are possible biochem-ical explanations of this difference (Thr308 phosphoryla-tion depends on the PDK1 kinase, whereas Ser473 phos-phorylation depends on mTOR) it is feasible that this

difference is simply linked to the reduced efficiency of p-AKT308 antibody (data not shown).

Deregulated activation of the AKT/mTOR cascade isinvolved in many human malignancies and its pharmaco-logic inhibitionwith selective drugs such as rapamycin or itsderivatives has shown efficacy in cancer treatment (15).Ourdatamayprovide the basis for future studies aimedat testingmTOR inhibitors for the treatment of metastatic MTC andmay also suggest suitable biomarkers to monitor the bio-chemical consequences of mTOR inhibitors in MTC.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: A. Tamburrino, J.S. Gutkind, M. SantoroDevelopment of methodology: A. Tamburrino, M. Raffeld, L. XiAcquisitionofdata (provided animals, acquired andmanagedpatients,provided facilities, etc.): R.D. Chernock, M. Raffeld, L. Xi, J.S. Gutkind, J.F.MoleyAnalysis and interpretation of data (e.g., statistical analysis, biosta-tistics, computational analysis): A. Tamburrino, A.A. Molinolo, M. Raf-feld, L. Xi, J.S. Gutkind, M. Santoro, S.A. WellsWriting, review, and/or revision of the manuscript: A. Tamburrino, A.A.Molinolo, R.D. Chernock, M. Raffeld, L. Xi, J.S. Gutkind, J.F. Moley, M.Santoro, S.A. WellsAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): A. Tamburrino, P. Salerno, L. Xi, S.A. WellsStudy supervision: J.S. Gutkind, M. SantoroExecution of the immunohistochemistry and in vitro and in vivoexperiments: A. TamburrinoDNA sequencing: P. Salerno

AcknowledgmentsThe authors thank Dr. Robert Gagel for the MZ-CRC-1 cell line and

AstraZeneca Inc. for providing us vandetanib.

Grant SupportThe studywas supported in part by a grant from the Italian Association for

Cancer Research (AIRC) to M. Santoro.The costs of publication of this article were defrayed in part by the

payment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

ReceivedOctober 21, 2011; revisedMarch 21, 2012; acceptedMay 4, 2012;published online July 2, 2012.

References1. SchlumbergerM, Carlomagno F, Baudin E, Bidart Jm, SantoroM. New

therapeutic approaches to treat medullary thyroid carcinoma. Nat ClinPract Endocrinol Metab 2008;4:22–32.

2. LiVolsi VA. C cell hyperplasia/neoplasia. J Clin Endocrinol Metab1997;82:39–41.

3. Skinner MA,Moley JA, DilleyWG, Owzar K, Debenedetti MK,Wells SAJr. Prophylactic thyroidectomy in multiple endocrine neoplasia type2A. N Engl J Med 2005;353:1105–13.

4. MachensA,Dralle H. Prophylactic thyroidectomy inRETcarriers at riskfor hereditary medullary thyroid cancer. Thyroid 2009;19:551–4.

5. Wells SA Jr, Santoro M. Targeting the RET pathway in thyroid cancer.Clin Cancer Res 2009;15:7119–23.

6. Fagin JA, Tuttle RM, Pfister DG. Harvesting the low-hanging fruit:kinase inhibitors for therapy of advanced medullary and nonmedullarythyroid cancer. J Clin Endocrinol Metab 2010;95:2621–4.

7. Sherman SI. Targeted therapy of thyroid cancer. Biochem Pharmacol2010;80:592–601.

8. Wells SA Jr, Gosnell JE, Gagel RF, Moley J, Pfister D, Sosa JA, et al.Vandetanib for the treatment of patients with locally advanced ormetastatic hereditary medullary thyroid cancer. J Clin Oncol2010;28:767–72.

9. Elisei R, Cosci B, Romei C, Bottici V, Renzini G, Molinaro E, et al.Prognostic significance of somatic RET oncogene mutations in spo-radic medullary thyroid cancer: a 10-year follow-up study. J ClinEndocrinol Metab 2008;93:682–7.

10. Elisei R, Romei C, Cosci B, Agate L, Bottici V, Molinaro E, et al. RETgenetic screening in patients with medullary thyroid cancer and theirrelatives: experience with 807 individuals at one center. J Clin Endo-crinol Metab 2007;92:4725–9.

11. Moura MM, Cavaco BM, Pinto AE, Leite V. High prevalence of RASmutations in RET-negative sporadic medullary thyroid carcinomas. JClin Endocrinol Metab 2011;96:E863–8.

12. HayN, SonenbergN.UpstreamanddownstreamofmTOR.GenesDev2004;18:1926–45.

Figure 5. Effects of rapamycin treatment of MZ-CRC-1 induced tumorxenografts. Mice (n ¼ 12) were injected with MZ-CRC-1 cells in bothflanks and when tumors appeared they were randomly assigned toreceive i.p. rapamycin (5 mg/kg/d) or vehicle. Tumor volume wasmeasured by calipers twice per week and reported as average value of 12tumors for each time point with SDs (��, P < 0.01). Inset, BrdUrdincorporation. Two hours before sacrifice, mice were given anintraperitoneal injection of BrdUrd. Animals were euthanized, and tissuewas fixed in 4% paraformaldehyde and embedded in paraffin for BrdUrdimmunohistochemistry. Values correspond to average percentage ofBrdUrd-positive nuclei (�SEM) in one slide each for 12 tumors(rapamycin) and 12 tumors (vehicle).

pS6 Phosphorylation in Metastatic MTC

www.aacrjournals.org Clin Cancer Res; 18(13) July 1, 2012 3539

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 9: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

13. Schubbert S, Shannon K, Bollag G. Hyperactive Ras in developmentaldisorders and cancer. Nat Rev Cancer 2007;7:295–308.

14. Cerrato A, De Falco V, Santoro M. Molecular genetics of medullarythyroid carcinoma: the quest for novel therapeutic targets. J MolEndocrinol 2009;43:143–55.

15. Foster KG, Fingar DC. Mammalian target of rapamycin (mTOR): con-ducting the cellular signaling symphony. J Biol Chem 2010;285:14071–7.

16. Zhou H, Luo Y, Huang S. Updates of mTOR inhibitors. AnticancerAgents Med Chem 2010;10:571–81.

17. Cooley LD, Elder FF, Knuth A, Gagel RF. Cytogenetic characterizationof three human and three rat medullary thyroid carcinoma cell lines.Cancer Genet Cytogenet 1995;80:138–49.

18. Vitagliano D, De Falco V, Tamburrino A, Coluzzi S, Troncone G,Chiappetta G, et al. The tyrosine kinase inhibitor ZD6474 blocksproliferation of RET mutant medullary thyroid carcinoma cells. EndocrRelat Cancer 2011;18:1–11.

19. de Groot JW, Links TP, Plukker JT, Lips CJ, Hofstra RM. RET as adiagnostic and therapeutic target in sporadic and hereditary endocrinetumors. Endocr Rev 2006;27:535–60.

20. Ma L, Chen Z, Erdjument-Bromage H, Tempst P, Pandolfi PP. Phos-phorylation and functional inactivation of TSC2 by Erk implications fortuberous sclerosis and cancer pathogenesis. Cell 2005;121:179–93.

21. Rapa I, Saggiorato E, Giachino D, Palestini N, Orlandi F, Papotti M,et al. Mammalian target of rapamycin pathway activation is associated

to RET mutation status in medullary thyroid carcinoma. J Clin Endo-crinol Metab 2011;96:2146–53.

22. Grozinsky-GlasbergS,RubinfeldH,NordenbergY,GorshteinA,PraissM, Kendler E, et al. The rapamycin-derivative RAD001 (everolimus)inhibits cell viability and interactswith theAkt-mTOR-p70S6Kpathwayin human medullary thyroid carcinoma cells. Mol Cell Endocrinol2010;315:87–94.

23. Kurokawa K, Iwashita T, Murakami H, Hayashi H, Kawai K, TakahashiM. Identification of SNT/FRS2 docking site on RET receptor tyrosinekinase and its role for signal transduction. Oncogene 2001;20:1929–38.

24. Melillo RM, Carlomagno F, De Vita G, Formisano P, Vecchio G,Fusco A, et al. The insulin receptor substrate (IRS)-1 recruitsphosphatidylinositol 3-kinase to Ret: evidence for a competitionbetween Shc and IRS-1 for the binding to Ret. Oncogene 2001;20:209–18.

25. Melillo RM, Santoro M, Ong SH, Billaud M, Fusco A, Hadari YR, et al.Docking protein FRS2 links the protein tyrosine kinase RET and itsoncogenic forms with the mitogen-activated protein kinase signalingcascade. Mol Cell Biol 2001;21:4177–87.

26. Encinas M, Tansey MG, Tsui-Pierchala BA, Comella JX, Milbrandt J,Johnson EM Jr. c-Src is required for glial cell line-derived neurotrophicfactor (GDNF) family ligand-mediated neuronal survival via a phos-phatidylinositol-3 kinase (PI-3K)-dependent pathway. J Neurosci2001;21:1464–72.

Tamburrino et al.

Clin Cancer Res; 18(13) July 1, 2012 Clinical Cancer Research3540

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Page 10: ActivationofthemTORPathwayinPrimaryMedullaryThyroid ... · S6, and the eukaryotic translation initiation factor 4E– binding protein 1 (4E-BP1; ref. 12). mTOR has received considerable

2012;18:3532-3540. Clin Cancer Res   Anna Tamburrino, Alfredo A. Molinolo, Paolo Salerno, et al.   Carcinoma and Lymph Node MetastasesActivation of the mTOR Pathway in Primary Medullary Thyroid

  Updated version

  http://clincancerres.aacrjournals.org/content/18/13/3532

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2012/06/25/18.13.3532.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/18/13/3532.full#ref-list-1

This article cites 26 articles, 8 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/18/13/3532.full#related-urls

This article has been cited by 5 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/18/13/3532To request permission to re-use all or part of this article, use this link

on June 9, 2020. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from