13
Small Molecule Therapeutics Delineating the mTOR Kinase Pathway Using a Dual TORC1/2 Inhibitor, AZD8055, in Multiple Myeloma Diana Cirstea 1,2 , Loredana Santo 1 , Teru Hideshima 2 , Homare Eda 1 , Yuko Mishima 1 , Neeharika Nemani 1 , Anuj Mahindra 1 , Andrew Yee 1 , Gullu Gorgun 2 , Yiguo Hu 2 , Hiroto Ohguchi 2 , Rikio Suzuki 2 , Francesca Cottini 2 , Sylvie M. Guichard 3 , Kenneth C. Anderson 2 , and Noopur Raje 1,2 Abstract Despite promising preclinical results with mTOR kinase inhibitors in multiple myeloma, resistance to these drugs may arise via feedback activation loops. This concern is especially true for insulin-like growth factor 1 receptor (IGF1R), because IGF1R signaling is downregulated by multiple AKT and mTOR feedback mechanisms. We have tested this hypothesis in multiple myeloma using the novel selective mTOR kinase inhibitor AZD8055. We evaluated p-mTOR S 2481 as the readout for mTORC2/Akt activity in multiple myeloma cells in the context of mTOR inhibition via AZD8055 or rapamycin. We next validated AZD8055 inhibition of mTORC1 and mTORC2 functions in multiple myeloma cells alone or in culture with bone marrow stroma cells and growth factors. Unlike rapamycin, AZD8055 resulted in apoptosis of multiple myeloma cells. AZD8055 treatment, however, induced upregulation of IGF1R phosphorylation in p-Akt S 473 –expressing multiple myeloma cell lines. Furthermore, exposure of AZD8055-treated cells to IGF1 induced p-Akt S 473 and rescued multiple myeloma cells from apoptosis despite mTOR kinase inhibition and TORC2/Akt blockage. The addition of blocking IGF1R antibody resulted in reversing this effect and increased AZD8055-induced apoptosis. Our study suggests that combination treatment with AZD8055 and IGF1R-blocking agents is a promising strategy in multiple myeloma with potential IGF1R/Akt signaling–mediated survival. Mol Cancer Ther; 13(11); 2489–500. Ó2014 AACR. Introduction Multiple myeloma is the second most common hema- tologic malignancy that is characterized by clonal prolif- eration of malignant plasma cells in the bone marrow (BM) microenvironment. Despite significant develop- ment of novel drugs in the last decade, multiple myeloma still remains incurable in the majority of patients (1). Current opinion is that molecularly informed targeted combination therapies will be required for long-term disease control (2). Aberrant activation of the PI3K–protein kinase B (PKB/ AKT) signaling in myeloma cells in the context of BM and cytokines led to the prediction that mTOR, a key protein of the PI3K/Akt pathway, may be a useful target in multiple myeloma. The serine/threonine kinase mTOR interacts with several proteins to form two distinct complexes named mTOR complex 1 (mTORC1) and 2 (mTORC2). mTORC1 phosphorylates p70 ribosomal protein S6 kinase (P70S6K) and translation initiation regulator 4E binding protein (4E-BP1), and regulates protein translation and cell size; mTORC2 phosphorylates Akt and SGK1, and regu- lates survival and cytoskeletal organization (3). mTOR is regulated via phosphorylation (4). There are several char- acterized phosphorylation sites in mTOR. T 2446 is phos- phorylated by adenosine monophosphate kinase and is regulated by nutrient availability. S 2448 is phosphorylated by P70S6K, reflecting the amino acid and nutrient status. S 2481 is the autophosphorylation site, which directly repre- sents intrinsic mTOR-specific catalytic activity (5). Initial studies targeting mTOR in multiple myeloma have focused on the inhibition of mTORC1 with rapamy- cin (6–8). The observed mTORC2 activation of Akt fol- lowing rapamycin treatment prompted the rapid devel- opment of mTOR kinase inhibitors (9). On the basis of recent preclinical testing of mTOR kinase in multiple myeloma (10), it was predicted that reduction in phos- phorylated (p) 4E-BP1 and p-Akt, both refractory to rapa- mycin, may provide preliminary evidence of mTOR kinase target inhibition and may overcome rapamycin- related resistance mechanisms. 1 MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts. 2 LeBow Institute for Myeloma Therapeu- tics and Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Dana-Farber Cancer Institute, Boston, Massachusetts. 3 AstraZeneca, Alderley Park, Maccleseld, United Kingdom. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). D. Cirstea and L. Santo contributed equally to this article. Current address for A. Mahindra: University of California San Francisco, Box 0324, UCSF, San Francisco, CA 94143-0324. Corresponding Author: Noopur Raje, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA 02114. Phone: 617-726-0711; Fax: 617-724-6801; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-14-0147 Ó2014 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 2489 on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

DelineatingthemTORKinasePathwayUsingaDualTORC1/2 ......kinase inhibitors have been proposed based on preclin-ical data (4). These include TORC1 and TORC2 down-stream targets, specifically

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

  • Small Molecule Therapeutics

    Delineating themTORKinasePathwayUsingaDual TORC1/2Inhibitor, AZD8055, in Multiple Myeloma

    Diana Cirstea1,2, Loredana Santo1, Teru Hideshima2, Homare Eda1, Yuko Mishima1, Neeharika Nemani1,Anuj Mahindra1, Andrew Yee1, Gullu Gorgun2, Yiguo Hu2, Hiroto Ohguchi2, Rikio Suzuki2, Francesca Cottini2,Sylvie M. Guichard3, Kenneth C. Anderson2, and Noopur Raje1,2

    AbstractDespite promising preclinical results with mTOR kinase inhibitors in multiple myeloma, resistance to these

    drugs may arise via feedback activation loops. This concern is especially true for insulin-like growth factor

    1 receptor (IGF1R), because IGF1R signaling is downregulated by multiple AKT and mTOR feedback

    mechanisms. We have tested this hypothesis in multiple myeloma using the novel selective mTOR kinase

    inhibitorAZD8055.We evaluatedp-mTORS2481 as the readout formTORC2/Akt activity inmultiplemyeloma

    cells in the context of mTOR inhibition via AZD8055 or rapamycin. We next validated AZD8055 inhibition of

    mTORC1 andmTORC2 functions inmultiplemyeloma cells alone or in culturewith bonemarrow stroma cells

    and growth factors. Unlike rapamycin, AZD8055 resulted in apoptosis of multiple myeloma cells. AZD8055

    treatment, however, induced upregulation of IGF1R phosphorylation in p-Akt S473–expressing multiple

    myeloma cell lines. Furthermore, exposure of AZD8055-treated cells to IGF1 induced p-Akt S473 and rescued

    multiple myeloma cells from apoptosis despite mTOR kinase inhibition and TORC2/Akt blockage. The

    addition of blocking IGF1R antibody resulted in reversing this effect and increased AZD8055-induced

    apoptosis. Our study suggests that combination treatment with AZD8055 and IGF1R-blocking agents is a

    promising strategy in multiple myeloma with potential IGF1R/Akt signaling–mediated survival. Mol Cancer

    Ther; 13(11); 2489–500. �2014 AACR.

    IntroductionMultiple myeloma is the second most common hema-

    tologic malignancy that is characterized by clonal prolif-eration of malignant plasma cells in the bone marrow(BM) microenvironment. Despite significant develop-ment of novel drugs in the last decade, multiple myelomastill remains incurable in the majority of patients (1).Current opinion is that molecularly informed targetedcombination therapies will be required for long-termdisease control (2).Aberrant activation of the PI3K–protein kinase B (PKB/

    AKT) signaling in myeloma cells in the context of BM and

    cytokines led to the prediction thatmTOR, a key protein ofthe PI3K/Akt pathway, may be a useful target in multiplemyeloma. The serine/threonine kinase mTOR interactswith several proteins to form two distinct complexesnamed mTOR complex 1 (mTORC1) and 2 (mTORC2).mTORC1 phosphorylates p70 ribosomal protein S6 kinase(P70S6K) and translation initiation regulator 4E bindingprotein (4E-BP1), and regulates protein translation and cellsize; mTORC2 phosphorylates Akt and SGK1, and regu-lates survival and cytoskeletal organization (3). mTOR isregulated via phosphorylation (4). There are several char-acterized phosphorylation sites in mTOR. T2446 is phos-phorylated by adenosine monophosphate kinase and isregulated by nutrient availability. S2448 is phosphorylatedby P70S6K, reflecting the amino acid and nutrient status.S2481 is the autophosphorylation site, which directly repre-sents intrinsic mTOR-specific catalytic activity (5).

    Initial studies targeting mTOR in multiple myelomahave focused on the inhibition of mTORC1 with rapamy-cin (6–8). The observed mTORC2 activation of Akt fol-lowing rapamycin treatment prompted the rapid devel-opment of mTOR kinase inhibitors (9). On the basis ofrecent preclinical testing of mTOR kinase in multiplemyeloma (10), it was predicted that reduction in phos-phorylated (p) 4E-BP1 and p-Akt, both refractory to rapa-mycin, may provide preliminary evidence of mTORkinase target inhibition and may overcome rapamycin-related resistance mechanisms.

    1MGH Cancer Center, Massachusetts General Hospital, Harvard MedicalSchool, Boston, Massachusetts. 2LeBow Institute for Myeloma Therapeu-tics and Jerome Lipper Center for Multiple Myeloma Research, HarvardMedical School, Dana-Farber Cancer Institute, Boston, Massachusetts.3AstraZeneca, Alderley Park, Macclesfield, United Kingdom.

    Note: Supplementary data for this article are available at Molecular CancerTherapeutics Online (http://mct.aacrjournals.org/).

    D. Cirstea and L. Santo contributed equally to this article.

    Current address for A. Mahindra: University of California San Francisco,Box 0324, UCSF, San Francisco, CA 94143-0324.

    Corresponding Author: Noopur Raje, Massachusetts General HospitalCancer Center, 55 Fruit Street, Boston, MA 02114. Phone: 617-726-0711;Fax: 617-724-6801; E-mail: [email protected]

    doi: 10.1158/1535-7163.MCT-14-0147

    �2014 American Association for Cancer Research.

    MolecularCancer

    Therapeutics

    www.aacrjournals.org 2489

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • However, cumulative evidence suggests that Akt acti-vation may occur even in the context of mTOR kinaseinhibition and blockage of TORC2/Akt signaling as aconsequence of disinhibition of feedback signaling (11).Specifically, relief of feedback inhibition of receptor tyro-sine kinases [RTK; e.g., EGFR, HER2, -3, -4, insulin-likegrowth factor 1 receptor (IGF1R), insulin receptor, andFGFR1-3] leading to subsequent PI3K activation and anmTOR-independent rephosphorylation of AKT at T308 issufficient to reactivate AKT signaling (12). Therefore, thelevels of p-Akt S473 in tumors treated with mTOR kinaseinhibitors may be misleading as a biomarker of eitherresponse or resistance.

    To date, suggested biomarkers of response to mTORkinase inhibitors have been proposed based on preclin-ical data (4). These include TORC1 and TORC2 down-stream targets, specifically the phosphorylated forms ofP70S6K, 4E-BP1, and Akt. Although evaluating multiplemarkers may offer a more robust evaluation of theinhibition of mTOR signaling, the baseline variabilitybetween mTORC1 and mTORC2 outputs in myelomatumors may present an obstacle in defining the mTORprofile of myeloma tumors sensitive to mTOR kinaseinhibitors. Moreover, pathway inhibition may not be auseful marker of response because different compo-nents of downstream signaling have differing thresh-olds for inhibition. Recent studies have proposed thatp-mTOR S2481 can be used to monitor mTOR kinaseinhibition. Moreover, it has been suggested that thisphosphosite is a better marker for mTORC2 activitythan Akt Ser473 phosphorylation (13, 14).

    We have now tested these assertions in multiple myelo-ma using the novel selective mTOR kinase inhibitorAZD8055 (15). Initially, we evaluated p-mTOR S2481 as thereadout for mTORC2/Akt activity in multiple myelomacells alone and in the context of mTOR inhibition viaAZD8055or rapamycin.We found thatAZD8055 treatmentinduced upregulation of IGF1R in multiple myeloma celllineswith constitutiveAkt activation, and that IGF1/IGF1RsignalingmediatedAktupregulationdespiteTORC2block-age. Our findings suggest that IGF1/IGF1R/Akt signalingmay bypass mTORC2 when mediating Akt activation,which may reduce apoptosis induced by mTOR kinaseinhibition. We believe the mTOR/Akt and IGF1R/Aktpathways cross-regulation might be of particular interestin multiple myeloma tumors with IGF1-enabled Akt acti-vation.Abetterunderstandingof the feedbackmechanismsresponsible for Akt activation inmultiple myelomawith orwithout stimulatory signals will complement the designof clinical trials for mTOR inhibitors either alone or incombination for patients with multiple myeloma.

    Materials and MethodsCell culture and reagents

    Humanmyeloma cell lines. Dex-sensitiveMM.1S and-resistantMM.1R cellswere provided byDr. StevenRosen(Northwestern University, Evanston, IL). U266 andRPMI8226 cell lines were obtained from the ATCC. Doxo-

    rubicin-resistant RPMI-DOX40 and Melphalan-resistantRPMI-LR5 cells were kindly provided by Dr. WilliamDalton (Lee Moffitt Cancer Center, Tampa, FL). OPM1and OPM2 plasma cell leukemia cell lines were kindlyprovided by Dr. Edward Thompson (University of TexasMedical Branch, Galveston, TX). IL6-dependent INA-6cell line was provided by Dr. Renate Burger (Universityof Kiel, Germany). These lines were characterized byshort tandem repeat profiling and compared with theknown ATCC database and German Collection of Micro-organismsandCellCulturesdatabases. The cell lineswerepassaged for fewer than 6 months following receipt orresuscitation from frozen stocks. All cell lines were cul-tured in RPMI-1640 containing 10% FBS (Sigma ChemicalCo.), 2 mmol/L L-glutamine, 100 U/mL penicillin, 100mg/mL streptomycin (GIBCO), with 2.5 and 5 ng/mL ofrecombinant IL6 only in INA-6 and ANBL-6 cells, respec-tively. Patient multiple myeloma primary tumor cellswere obtained from BM aspirates in accordance with theDeclaration of Helsinki and with the approval of theInstitutional Review Board of Massachusetts GeneralHospital. BM mononuclear cells were separated usingFicoll–Hipaque density sedimentation, and plasma cellswere purified by positive selection with anti-CD138 mag-netic activated cell separation microbeads (Miltenyi Bio-tec). BM stroma cells (BMSC) were generated from BMspecimens from patients with multiple myeloma asdescribed previously (16). BMSCs (103 cells/well) wereincubated in 96-well culture plates for 24 hours, multiplemyeloma cells were then added to thewells (2� 104 cells/well) and incubated with media alone, or with increasingconcentrations of AZD8055 or rapamycin for the specifiedtimes at 37�C. Blood samples collected from healthyvolunteers were processed by Ficoll–Hipaque gradientto obtain peripheral blood mononuclear cells (PBMC).

    AZD8055. Selective and orally bioavailableATP-com-petitive mTOR kinase inhibitor was obtained from Astra-Zeneca. For in vitro studies, AZD8055 was prepared as 10mmol/L stock solution inDMSO and stored at�20�Canddiluted in culture medium (1–200 nmol/L) immediatelybefore use. For studies inmice, AZD8055was prepared asa suspension in sterile (autoclaved) vehicle 0.5% hydro-xpropylmethylcellulose (ref. 56336; Fluka) þ 0.1% poly-sorbate 80 (ref. 93780, Tween 80; Fluka), sterilized byautoclaving at 121�C for 20 minutes at 30 psi (1.5 Bar).AZD8055 was administered by oral gavage (0.1 mL/10 gof body weight) once or twice daily.

    Rapamycin. Rapamycin was obtained from Calbio-chem (EMD Millipore).

    Akti-1/2. Akt 1/2 selective inhibitor was purchasedfrom Calbiochem (EMD Millipore).

    Cell viability and proliferation assaysColorimetric assay. Colorimetric assayswere done to

    assay drug activity. Cell cultures were pulsed with 10 mLof 5 mg/mL MTT (Chemicon International, Inc.) to eachwell, followed by incubation at 37�C for 4 hours, andaddition of 100mL isopropanolwith 0.04HCl.Absorbance

    Cirstea et al.

    Mol Cancer Ther; 13(11) November 2014 Molecular Cancer Therapeutics2490

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • readings at awavelength of 570 nm (with correction usingreadings at 630 nm) were taken on a spectrophotometer(Molecular Devices Corp.).Proliferation assay. DNAsynthesiswasmeasuredby

    tritiated thymidine uptake (3H-TdR; Perkin-Elmer) aspreviously described (8). Briefly, MM.1S cells (2–3 �103 per well) were incubated in 96-well culture platesalone or in culture with BMSCs, IL6 (10 ng/mL), or IGF1(50 ng/mL) in the presence of media or varying concen-trations of AZD8055 or rapamycin for 48 hours at 37�C.

    ImmunoblottingMultiple myeloma cells were cultured in media or

    with indicated concentrations of AZD8055 or rapamycin,harvested, and whole-cell lysates were subjected toSDS-PAGE, and transferred to nitrocellulose membrane(Bio-Rad Laboratories), as described previously (8).Immunoblotting was carried using specific antibodies:anti–p-mTOR S2481, -mTOR S2448, -mTOR, –p-P70S6KT389, –p-P70S6K T421/S424, -P70S6K, –p-4E-BP1 T37/46,–p-4E-BP1 S65, –p-Akt S473, -Akt, –p-NDRG1 S330, -LC3B,-cleaved caspase-3, -7, -8, -9, -PARP, –p-IGF1R T1135/1136,and -IGF1R b (Cell Signaling Technology). Same lysateshave been used for blotting total proteins and their phos-phorylated forms. Blots were reprobedwith anti-GAPDHantibody (Cell Signaling Technology) for equal proteincontrol when there was unequal expression of total pro-tein.Antigen–antibody complexesweredetected byusingenhanced chemiluminescence (Amersham).

    Protein synthesis assayProtein synthesis rate was measured using The CytoX-

    Red Cell Proliferation/Cytotoxicity Assay Kit (EpigentekGroup Inc.) according to the manufacturer’s protocol.MM.1S cells (2 � 103 per well) were incubated in 96-wellculture plates alone or with AZD8055 (50, 100, and 200nmol/L) or rapamycin (100 nmol/L). After 24- or 48-hourincubation, the growth media were removed, cellswashed with PBS, and 50 mL of indicator solution addedto eachwell for 10 to 15minutes at room temperature. Theplate was washed, 100 mL of dissolving solution added toeachwell, and optical density (O.D.) wasmeasured at 570nanometers (nm).

    Detection of apoptosis by Annexin V/PropidiumIodide stainingDetection of early apoptotic cells was performed with

    the Annexin V–PI (Annexin/PI) detection Kit (Immuno-tech/Beckman Coulter). Briefly, MM.1S cells (1 � 106)were exposed for various time points to AZD8055 orrapamycin. Cells were washed and then incubated in thedark at room temperature with Annexin V–FITC and PIfor 15 minutes. Annexin Vþ/PIþ apoptotic cells wereenumerated using the Epics flow cytometer.

    Multiple myeloma xenograft murine modelThe in vivo anti–multiple myeloma activity of AZD8055

    was evaluated in a previously established multiple mye-

    loma xenograftmodel (8). CB-17 SCIDmice obtained fromCharles River Laboratories were subjected to animalstudies according to the protocols approved by the Ani-mal Ethics Committee. Forty 5- to 6-week-old male micewere irradiated [2 Gy (200 rad)] using cesium 137[(137Cs)-irradiator source]; 24 hours after irradiation,2.5 � 106 MM.1S cells suspended in 100 mL of RPMImedium were inoculated subcutaneously in the upperback. When tumors were measurable with a tumor vol-ume of approximately 150 to 200 mm3, mice were ran-domly assigned into three cohorts receiving daily gavagesfor 5 days per week for 4 consecutive weeks with eitherAZD8055 5 mg/kg (10 mice), 10 mg/kg (10 mice), orcontrol vehicle alone (10 mice) on the same schedule.Animals were monitored for body weight and tumorvolume by caliper measurements every day. Tumorvolume was estimated using the following formula:1/2 � (length) � (width)2. Animals were euthanized inaccordance with institutional guidelines by CO2 inha-lation in the event of tumor size >2 cm or major com-promise in their quality of life, due to tumor ulceration.Survival was evaluated from the first day of treatmentuntil death. Tumor growth was evaluated using calipermeasurements from the first day of treatment until dayof first sacrifice. Percentage tumor growth inhibition(TGI) was calculated as TGI on day X ¼ 100 � T/C%on day X.

    Statistical analysisAll in vitro experiments were performed in triplicate

    and repeated at least 3 times; a representative experimentwas selected for figures. Statistical significance of differ-ences was determined using the Student t test, withminimal level of significance (P < 0.05). In vivo statisticaltests were performed using the Student t test. Overallsurvival (OS) was measured using the Kaplan–Meiermethod, and results are presented as themedian OS, with95% confidence intervals.

    ResultsmTOR/Akt signaling in multiple myeloma cells:p-mTOR S2481 as a marker for mTORC2-phosphorylatable Akt S473

    We first examined whether autophosphorylation ofmTOR on S2481 can be used as readout for mTORC2/Aktsignaling in multiple myeloma cells. We evaluated thebaseline phosphorylation of mTOR on S2481 in multiplemyeloma cell lines (Fig. 1A).Normally,mTORC1 containsmainlymTORphosphorylated at S2448, whereasmTORC2contains predominantly mTOR phosphorylated at S2481

    (13). Through Western blot analysis, we found thatp-mTORS2481waspresent only inMM.1S,MM.1R,OPM1,OPM2, and U266 cells, whereas p-mTOR S2448 wasexpressed in all multiple myeloma cell lines. We nextcompared the expression of mTOR phosphorylation onS2448 and S2481 with TORC1 and TORC2 readouts, respec-tively. The best-characterized mTORC1 readouts are

    Targeting mTOR Kinase/IGF1R Feedback in Multiple Myeloma

    www.aacrjournals.org Mol Cancer Ther; 13(11) November 2014 2491

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • P70S6K and 4E-BP, whereas Akt is a commonly usedreadout formTORC2activity.Ourdata show that baselineexpression of p-mTORS2448 correlatedwithTORC1-phos-phorylatable P70S6K and 4E-BP, and expression of p-

    mTOR S2481 correlated with TORC2-phosphorylatableAkt (S473) across all multiple myeloma cell lines, exceptU266 inwhich p-mTORS2481 is highly expressed,whereasp-AKT S473 is not significantly expressed.

    Figure 1. mTOR/Akt signaling in multiple myeloma cells: p-mTOR S2481 as a marker for mTORC2-phosphorylatable Akt S473. A, baseline mTOR profile inmultiple myeloma cell lines. Multiple myeloma cells grown in culture media alone were lysed and mTOR/TORC1/TORC2-related protein expressionanalyzed by Western blotting. B, effect of serum starvation/readdition on mTOR activity in multiple myeloma cells. MM.1S, OPM1, and RPMI multiplemyeloma cells were cultured in regular (10% FBS) or serum-starved media for 72 hours; next, regular media were added to 72-hour–starved cells for 4, 8, 24,48, or 72 hours, and whole-cell lysates subjected to Western blotting for p-mTOR S2481/S2448, mTOR, p-P70S6K T389, P70S6K, p-Akt S473, and Aktexpression. GAPDH was used for loading control. C, Akt regulates p-mTOR S2481. MM.1S, OPM1, and RPMI cells exposed to media alone or with Akti-1/2(5 mmol/L) for 30 minutes, 1 hour, 3 hours, or 6 hours were analyzed by Western blotting for the expression of p-Akt S473, Akt, p-mTOR S2481, and mTOR.D, p-mTOR S2481 correlates with p-Akt S473 expression in CD138þ multiple myeloma primary tumor cells. CD138þ primary tumor cells from patientswith multiple myeloma and PBMCs from healthy donors were subjected to Western blotting, and p-mTOR S2481, mTOR, p-Akt S473, Akt, and GAPDHexpression detected using specific antibodies.

    Cirstea et al.

    Mol Cancer Ther; 13(11) November 2014 Molecular Cancer Therapeutics2492

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • Because serumstarvation leads to auniformresponse inthemTORpathway (17), we next compared the kinetics ofS2448/S2481 phosphorylation of mTOR with p-Akt S473

    levels in multiple myeloma cell lines with relatively high(MM.1S and OPM1 cells) versus low (RPMI cells) consti-tutive expression of p-Akt S473 (Fig. 1B). MM.1S, OPM1,and RPMI cells cultured in serum-deprived media for 72hours and then stimulatedwith serum for increasing timepoints (4–72 hours) were subjected toWestern blotting. InRPMI cells, P-mTOR S2481 and p-Akt S473 followed similarkinetics of expressionduring serumwithdrawal and read-dition. In MM.1S cells, the initial induction of p-Akt S473

    occurred before p-mTOR S2481 induction, suggesting thateither low levels [not easily seen by Western blotting(WB)] of p-mTOR S2481 are sufficient to induce p-AKTS473 or that TORC2 is not the only candidate to phosphory-late Akt at S473 in this cell line. In OPM1 cells, p-mTORS2481 and p-Akt S473 coexpression was present even inserum-starved cells, consistent with PI3K/Akt pathwayconstitutive activation due to PTEN deletion.Because Akt plays a role in the regulation of mTOR, we

    next examinedwhether p-mTORS2481 is dependent on theAkt activation status (Fig. 1C). We found that reduced

    expression of p-Akt S473 via treatment with the Akt inhib-itor Akti-1/2 resulted in reducedmTOR phosphorylationat S2481 in MM.1S and RPMI, but not in OPM1 cells.

    Finally,we tested p-mTORS2481 and p-Akt S473 baselineexpression in CD138þ cells obtained from three patientswith multiple myeloma, and in PBMCs from five healthydonors (Fig. 1D). We found that p-mTOR S2481 expressioncorrelated with p-Akt S473 in both CD138þ and healthydonor mononuclear cells.

    AZD8055, an mTOR kinase inhibitor, inhibitsmultiple myeloma cell growth in vitro and in vivo

    We then evaluated the effect of targeting TORC1 andTORC2 in MM by using a novel selective mTOR kinaseinhibitor, AZD8055 (15). We first assessed the effects ofAZD8055 (25–200 nmol/L) treatment on cell number andmetabolic activity of multiple myeloma cell lines by MTTassay at 72hours.Wenoteddose-dependentdifferences ingrowth among cells with high versus low p-mTOR S2481/p-Akt S473 expression, with MM.1S, MM.1R, OPM1, andOPM2 cells being more sensitive to AZD8055 treatment(Fig. 2A). We also examined AZD8055 activity in freshlyisolated CD138þ tumor cells from patients with multiple

    Figure 2. AZD8055, an mTOR kinase inhibitor, shows in vitro and in vivo antimyeloma effect. A, AZD8055 decreases multiple myeloma viability. Multiplemyeloma cell lines were cultured in culture media or at varying concentrations of AZD8055 (0–200 nmol/L), and cytotoxicity evaluated by MTT assayat 72 hours; data,mean (�SD) of triplicate culture. B, cells of patientswithmultiplemyeloma exhibited differential sensitivity to AZD8055. Cells of patientswithmultiple myeloma isolated from BM by CD138-positive selection were cultured with or without AZD8055 (100 or 200 nmol/L) for 72 hours and viabilityassessed via MTT assay. Data, mean (�SD) of triplicate culture. C, AZD8055 was not toxic to PBMCs. Healthy donor PBMCs were incubated with orwithout increasing concentrations of AZD8055 (0–800 nmol/L) and viability assessed at 72 hours by MTT assay; data, mean (�SD) of triplicate culture.D, AZD8055 anti–multiple myeloma activity in vivo: tumor growth, host weight, and Kaplan–Meier survival curves in xenografted with MM.1S cells mice.Immunodeficient SCID-CB17 mice were inoculated subcutaneously MM.1S cells. Oral AZD8055 was administered daily 5 days per week for 4 consecutiveweeks either 5 mg/kg (10 mice), 10 mg/kg (10 mice), or control vehicle (10 mice) starting after the development of measurable tumor with a tumorvolume of approximately 150 to 200 cm3. AZD8055 treatment resulted in tumor growth inhibition and prolonged survival.

    Targeting mTOR Kinase/IGF1R Feedback in Multiple Myeloma

    www.aacrjournals.org Mol Cancer Ther; 13(11) November 2014 2493

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • myeloma. As evidenced by the MTT assays, AZD8055(100–200 nmol/L) treatment (72 hours) inhibited the pro-liferation of three of eight tested samples (Fig. 2B).Increased dosing of AZD8055 (0–800 nmol/L) did nottrigger significant cytotoxicity in healthy donors PBMCs(Fig. 2C). We next validated AZD8055’s anti–multiplemyeloma activity in vivo in theMM.1S cell xenograft SCIDmouse model (Fig. 2D). Treatment with AZD8055 signif-icantly suppressedmultiplemyeloma tumor growth,withmaximumTGI (%) noted at days 19 and 24 following startof treatment at 65% and 59% in the 10mg/kg and 5mg/kgtreated cohorts, respectively (control vs. treated, P < 0.05).AZD8055 treatment was also associated with improvedOS (control vs. treated, P < 0.05; Fig. 2D, left). No toxicdeaths occurred during this study: maximum percentageof body weight loss was observed on day 5 (1.3%) at 5mg/kg dosage schedule and on day 8 (6%) after 10mg/kgdosing, with weight recovery in the following 2 weekswith continued dosing (Fig. 2D).

    AZD8055 blocks p-mTOR S2481/p-Akt S473 signalingand displays higher antimyeloma effect comparedwith rapamycin

    We next examined how p-mTOR S2481/p-Akt S473

    expression correlates with response to mTOR-targetedtreatment in multiple myeloma cells. We compared p-mTOR S2481/p-Akt S473 response to AZD8055 and torapamycin treatment in MM.1S cells. Time course West-ern blot analysis ofMM.1S cells exposed to AZD8055 (100nmol/L) demonstrated inhibition of phosphorylation ofboth TORC1 and TORC2 substrates: P70S6K and 4E-BP1including the rapamycin-resistant T37/46—downstreamtargets of TORC1, as well as Akt and NDRG1—effectorsof TORC2 refractory to rapamycin (Fig. 3A). Unlike rapa-mycin, AZD8055 consistently inhibited mTOR phosphor-ylation at S2481 along with p-Akt S473, as evidenced byWestern blot studies in MM.1S, OPM1, and RPMI cells(Fig. 3B). These results reflected in a different response interm of cell viability when the multiple myeloma cellswere exposed to the same concentration of AZD8055 andrapamycin (AZD8055 vs. rapamycin MM1S P ¼ 0.0011,MM1R P ¼ 0.0001, OPM1 P ¼ 0.0001, OPM2 P < 0.0001,RPMI P¼ 0.0016, DOX-40 P¼ 0.0016, LR5 P¼ 0.0005, andINA-6 P ¼ 0.0034), as shown by MTT assay (Fig. 3C).AZD8055 was more potent than rapamycin and resultedin a greater impact on cell viability.

    We next evaluated the cellular effects of the AZD8055-mediated inhibition of mTORC1 and mTORC2 functionsinMM.1S cells. mTORC1 is a positive regulator of proteinsynthesis through both inhibition of autophagy andinduction of P70S6K/4E-BP1–mediated protein transla-tion. Our Western blot time course (0, 3, 6, 12, 24, and 48hours) analysis of LC3 I and LC3 II expression showedthat AZD8055 (100 nmol/L) induced a clear cleavage ofLC3BI into LC3BII. Such a clear effect was not observedafter rapamycin (100 nmol/L) treatment. Although pro-longed treatment with rapamycin increased the expres-sion of both LC3 I and LC3 II bands, prolonged treatment

    with AZD8055 reduced LC3 I at 3 hours and triggered thedisappearance of both bands by 24 hours (Fig. 3D, left).BothAZD8055 and rapamycin 24- and 48-hour treatmentsresulted in up to 80% inhibition of protein synthesis(AZD8055 50 nmol/L vs. rapamycin 100 nmol/L, 48hours, P ¼ 0.5; AZD8055 100 nmol/L vs. rapamycin 100nmol/L, 48 hours, P ¼ 0.16; AZD8055 200 nmol/L vs.rapamycin 100 nmol/L, 48 hours, P¼ 0.11; Fig. 3D, right).

    BecausemTORC2 is a positive regulator of cell survivalvia Akt-mediated inhibition of apoptosis, we examinedwhether AZD8055 could trigger apoptosis in multiplemyeloma cells. Consistent with mTORC2 inhibition,AZD8055, unlike rapamycin, induced apoptosis, as evi-denced by Western blot detection of caspase-7, -8, -9, -3,and PARP cleavage in a time-dependent fashion (Fig. 3E,left), and confirmed by increased population of apoptoticcells at 72-hour culture as detected by Annexin V/PIstaining (Fig. 3E, right).

    IGF1-induced Akt activation confers relativeresistance to AZD8055-mediated cytotoxicity

    Becausewe, and others, have reported on the role of theBM microenvironment in mTOR/Akt-mediated prolifer-ation and survival in multiple myeloma cells, we nextexamined AZD8055’s efficacy in multiple myeloma cellsin culture with BMSCs or growth factors such as IL6 andIGF1. We have previously shown that rapamycin alonewas unable to overcome the protective effects of BMSCsand growth factors (7, 8). We therefore evaluatedAZD8055’s activity next to rapamycin in MM.1S cultureswith or without BMSCs via [3H]-thymidine uptake at 48hours. AZD8055 resulted in greater antiproliferativeeffects as compared with rapamycin. AZD8055 treatmenthowever could not completely overcome BMSCs-medi-ated growth of MM.1S cells at 100 nmol/L dose (Fig. 4A).Because of the major role played by IL6 and IGF1 inmultiple myeloma cell survival triggered by BM micro-environment,wenext examinedAZD8055and rapamycintreatments ofMM.1S cells culturedwith orwithout IL6 (10ng/mL) or IGF1 (50 ng/mL). [3H]-thymidine uptake at 48hours demonstrated that AZD8055 treatment completelyabrogated IL6-mediated proliferation; meanwhile, IGF1conferred relative resistance to AZD8055 in a mannersimilar to BMSCs (Fig. 4B).

    Because recent evidence suggests that mTOR and Aktnegatively regulate IGF1R expression, and that IGF1Ractivity is reactivated by mTOR kinase inhibitors(12, 18, 19), we sought to determine whether AZD8055triggered IGF1R feedback activation inmultiplemyelomacells. We tested our hypothesis in two multiple myelomacell lineswith relatively high baselineAkt activity (MM.1Sand OPM1). Multiple myeloma cells were cultured for 6hours in control media, AZD8055 (100 nmol/L) or rapa-mycin (100 nmol/L), then stimulated with IGF1 (50ng/mL) for 15 minutes, and whole-cell lysates were sub-jected to Western blot analysis. We found that AZD8055alone induced the phosphorylation of tyrosine sites(Y1135/1136) in the activation loop of the IGF1R kinase

    Cirstea et al.

    Mol Cancer Ther; 13(11) November 2014 Molecular Cancer Therapeutics2494

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • Figure 3. AZD8055 inhibits cell growth by blocking p-mTOR S2481/p-AKT S473 signaling. A, AZD8055, an mTOR kinase inhibitor, induces dual inhibition ofTORC1/TORC2 signaling in multiple myeloma cells. MM.1S cells were incubatedwith AZD8055 (100 nmol/L) or rapamycin (100 nmol/L) for the specified timepoints, subjected to Western blot, and mTORC1/mTORC2 downstream targets expression was detected using specific antibodies. B, p-mTOR S2481

    is a marker of mTOR kinase inhibition in multiple myeloma cells. MM.1S, OPM1, and RPMI cells were incubated with AZD8055 (100 nmol/L) or rapamycin(100 nmol/L) for short time points (15 minutes to 6 hours). Cells were pelleted and subjected to Western blotting. p-mTOR S2481, mTOR, p-Akt S473, and Aktexpression was determined using specific antibodies. C, p-mTOR S2481/p-Akt S473 coexpression correlates with sensitivity to mTOR kinase inhibition inmultiple myeloma cell lines. Multiple myeloma cell lines were cultured in culture media or with AZD8055 (100 nmol/L) or rapamycin (100 nmol/L), andcytotoxicity was evaluated by MTT assay at 72 hours; data, mean (�SD) of triplicate culture. D, left, AZD8055 induced autophagy, consistent with TORC1inhibition. MM.1S cells were incubated with media, AZD8055 (100 nmol/L), or rapamycin (100 nmol/L) for 3, 6, 12, 24, or 48 hours, and cells analyzed forLC3B expression using Western blot. GAPDH expression served as a loading control. Right, AZD8055 blocked protein synthesis, a TORC1-relatedfunction. Protein synthesis was assayed in MM.1S cells cultured for 24 and 48 hours either in regular media or with increasing concentrations of AZD8055(50–200 nmol/L) or rapamycin (100 nmol/L). E, left, AZD8055 induced caspases activation, consistent with TORC2/Akt inhibition. MM.1S cells wereincubated with or without AZD8055 (100 nmol/L) or rapamycin (100 nmol/L) for 24 and 48 hours. Cleaved caspase-7, -8, -9, -3, and PARP were detectedvia WB using specific antibodies. Right, AZD8055 induced apoptosis. MM.1S cells were treated with AZD8055 (100 nmol/L) or rapamycin (100 nmol/L)for 48 and 72 hours. The rate of apoptosis was assessed by Annexin/PI flow cytometry analysis.

    Targeting mTOR Kinase/IGF1R Feedback in Multiple Myeloma

    www.aacrjournals.org Mol Cancer Ther; 13(11) November 2014 2495

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • Figure 4. IGF1-induced Aktactivation confers relativeresistance to AZD8055-mediatedcytotoxicity. A, BMSC-stimulatedMM.1S cells partially escapeAZD8055-mediated growthinhibition. MM.1S cells werecultured for 48 hours in controlmedia, AZD8055 (25–200 nmol/L),or rapamycin (10–100 nmol/L) inthe presence or absence ofBMSCs. Cell proliferation wasassessed by [3H]TdR assay; data,mean (�SD) of triplicate cultures.B, AZD8055-inhibited proliferationof MM.1S is partially rescued byIGF1. MM.1S cells were culturedfor 48 hours in control media,AZD8055 (25–200 nmol/L), orrapamycin (10–100 nmol/L) in thepresence or absence of IL6(10 ng/mL) or IGF1 (25 ng/mL). Cellproliferation was assessed by[3H]TdR assay; data, mean (�SD)of triplicate cultures. C, AZD8055treatment triggered upregulation ofthe tyrosine phosphorylation sitesin the activation loop of the IGF1Rkinase domain T1135/1136, whichassociated with activated p-AktS473 in multiple myeloma cellsdespite AZD8055 exposure.MM.1S and OPM1 cells werecultured for 6 hours in regularmedia, AZD8055 (100 nmol/L), orrapamycin (100 nmol/L). Cells werethen pelleted down andresuspended in PBS with orwithout IL6 (10 ng/mL) or IGF1 (50ng/mL) for 15 minutes. Expressionof p-IGF1 T1135/1136, IGF1Rb,p-mTOR S2481, p-mTOR S2448,mTOR, p-Akt S437, and Akt wasexamined via Western blot usingGAPDH for loading control. D, IGF1confers relative resistance toAZD8055-induced cytotoxicity ofmultiple myeloma. MM.1S andOPM1 multiple myeloma cellsincubated for 48 hours in mediaalone or AZD8055 (50, 100 nmol/L),with or without 50 ng/mL IGF1,were assessed for viability via MTTassay; data, mean (�SD) oftriplicate cultures.

    Cirstea et al.

    Mol Cancer Ther; 13(11) November 2014 Molecular Cancer Therapeutics2496

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • domain. IGF1 clearly upregulated Akt phosphorylationin multiple myeloma cells; however, it had no effect onmTOR Ser2481 phosphorylation. Moreover, AZD8055-trea-tedcells exposed to IGF1sustainedp-AktSer473 expression,whereas p-mTOR Ser2481 remained inhibited (Fig. 4C).Reactivation of IGF1R signaling in multiple myeloma

    cells in the context of mTOR kinase inhibitors suggeststhat multiple myeloma may survive in an IGF1-depen-dent fashion.We therefore next treatedMM.1S andOPM1cellswithAZD8055, in the presence or absence of IGF1 (50ng/mL). As evidenced by the MTT assay at 48 hours,multiple myeloma cells partially escaped AZD8055 cyto-toxicity (Fig. 4D).

    Blockage of IGF1R restores AZD8055-mediatedmultiple myeloma cytotoxicity

    We next determined whether blocking IGF1R would(i) prevent IGF1-mediated Akt reactivation in AZD8055-treated multiple myeloma cells, and (ii) reinstituteAZD8055’s anti–multiplemyeloma activity compromisedby IGF1. We performed Western blot analysis of MM.1Scells treated with AZD8055 (100 nmol/L) or anti–IGF1RAb (1 mg/mL), or the combination, with or without theaddition of 50 ng/mL IGF1 for 6 hours. Blockade of IGF1Rsignalingwith the anti–IGF1RAb alone resulted in partialreduction of Akt S473 phosphorylation. We confirmed theanti–IGF1R Ab–mediated downregulation of Akt activity

    Figure 5. Blockage of IGF1Rrestores AZD8055-mediatedmultiple myeloma cytotoxicity.A, IGF1R antibody prevents IGF1-mediated Akt reactivation inAZD8055-treated MM.1S cells.MM.1S cells were preincubated for1 hour with Iso-IgG Ab (1 mg/mL) oranti–IGF1R Ab (1 mg/mL) and thenexposed to 100 nmol/L AZD8055with or without 50 ng/mL IGF1 for6 hours. Expression of p-mTORS2481, mTOR, p-Akt S437, Akt,and p-FoxO1 T24/FoxO3a T32 wasexamined via Western blot usingGAPDH for loading control. B, thecombination of AZD8055 with theanti–IGF1R Ab restores AZD8055-mediated caspase cleavageimpaired by IGF1. MM.1S cellscultured for 24 hours with 100nmol/L AZD8055, 1 mg/mL anti–IGF1R Ab, or their combination,with or without 50 ng/mL IGF1,were subjected to Western blotanalysis for caspase-9 and -3expression. C, blockage of IGF1Rvia anti–IGF1R Ab prevents IGF1-mediated reduction in apoptosisproduced by AZD8055 treatment.Annexin/PI flow cytometry analysiswas performed to determine therate of apoptosis in MM.1S cellssubjected to longer treatment (72hours) with AZD8055 (100 nmol/L)with or without anti–IGF1R Abpretreatment, with or without IGF1(50 ng/mL). D, IGF1R blockage viaIGF1R Ab potentiates AZD8055activity in MM.1R and ANBL-6cells. MM.1R and ANBL-6 cellspreincubated for 1 hour with Iso-IgG Ab (1 mg/mL) or anti–IGF1R Ab(1 mg/mL) were exposed toAZD8055 (100 nmol/L) with orwithout IGF1 (50 ng/mL) for 72hours and viability assessed viaMTT assay; data, mean (�SD) oftriplicate cultures.

    Targeting mTOR Kinase/IGF1R Feedback in Multiple Myeloma

    www.aacrjournals.org Mol Cancer Ther; 13(11) November 2014 2497

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • by reduced p-FoxO1 T24/FoxO3a T32. Next, 50 ng/mLIGF1 partially rescued the phosphorylation of Akt S473/FoxO1 T24/FoxO3a T32 signaling in both AZD8055- andanti–IGF1R-treated cells, but not in the combination treat-ment. Notably, mTOR S2481 phosphorylation remainedunchanged either with IGF1 stimulation or with anti–IGF1R Ab treatment (Fig. 5A).

    We hypothesized that IGF1/IGF1R-mediated signalingcould rescue MM.1S cells from AZD8055-mediated apo-ptosis. Through Western blot analysis of MM.1S cellscultured for 24 hours with 100 nmol/L AZD8055 or 1mg/mL anti–IGF1R Ab, or the combination with or with-out 50 ng/mL IGF1, we found that IGF1 significantlyreduced caspase-3 cleavage induced by AZD8055 treat-ment in MM.1S. In contrast, the combination of AZD8055with the anti–IGF1R Ab increased the activation of cas-pases-9 and -3 (Fig. 5B).

    We therefore usedAnnexin/PI flow cytometry analysistodetermine the rate of apoptosis inMM.1S cells subjectedto longer treatment (72 h) with AZD8055 (100 nmol/L)with or without anti–IGF1R Ab pretreatment, with orwithout the addition of IGF1 (50 ng/mL). AZD8055 aloneinduced 56.5% apoptotic cells; the addition of anti–IGF1RAb increased the apoptotic population to 77.9%. Ashypothesized, exposure to IGF1 reduced AZD8055-ind-uced apoptosis from 56.5% to 15.1%, whereas blockage ofIGF1R via anti–IGF1R Ab partially rescued AZD8055-mediated apoptotic effect (Fig. 5C).

    To confirm our observation, we evaluated AZD8055and the anti–IGF1R Ab combined treatment in two othermultiplemyeloma cell lines,MM.1R andANBL-6.MM.1Rand ANBL-6 cells were preincubated for 1 hour with 1mg/mL Iso-IgG or 1 mg/mL anti–IGF1R Ab and thenculturedwithAZD8055 (0–100 nmol/L) or regularmedia,with or without 50 ng/mL IGF1. After 72-hour treatment,anti–IGF1R Ab alone reduced the cell numbers by 73%and 33% in MM.1R and ANBL-6 cells, respectively; next,anti–IGF1R Ab enhanced AZD8055 activity (85% cellnumber decrease in MM1R treated with AZD8055 vs.94% cell number decrease in MM1R treated withAZD8055 and anti–IGF1R Ab, 46% cell number decreasein ANBL-6 treated with AZD8055 vs. 66% cell numberdecrease in ANBL-6 treated with AZD8055 and anti–IGF1R Ab. P < 0.05) and reversed IGF1-mediated partialrescue of multiple myeloma cells fromAZD8055-inducedcytotoxicity (Fig. 5D). To confirm these findings, we sup-pressed the expression of IGF1R using siRNA inU266 celllines. Cells with IGF1R knockout showed 39% decrease inviability compared with scramble control, and the com-bination of IGF1R knock out and AZD8055 100 nmol/Lshowed a 78%decrease in viability comparedwith scram-ble control (Supplementary Fig. S1).

    DiscussionThe growing complexity of themTOR pathway, as well

    as emerging discoveries about its autoregulatorymechan-isms and activating feedback loops (4, 9), highlights the

    need for ongoing research of the mTOR pathway inmultiple myeloma. Our data aligned with previous find-ings in multiple myeloma (10) suggest that targetingmTOR kinase produces more effective mTOR pathwayinhibition than rapamycinwith amoredetrimental impacton multiple myeloma cells. Unlike rapamycin, AZD8055inhibition ofmTORC1 andmTORC2 functions resulted inapoptosis in multiple myeloma cells. However, we alsofound that mTOR kinase inhibition via AZD8055 inducedphosphorylation of IGF1R. Furthermore, exposure ofAZD8055-treated cells to IGF1 recovered p-Akt S473 andreduced apoptosis of multiple myeloma cells despitemTORkinase inhibition. In contrast, the addition of block-ing IGF1R antibody rescued AZD8055-induced apoptosisfrom IGF1-mediated multiple myeloma cell survival.

    Taken together, our findings indicate that p-Akt S473 isnot an accuratemarker formTORC2 inhibition inmultiplemyeloma cells. So far, several biomarkers have beendeveloped to monitor the effects of mTOR kinase inhibi-tors. These include measurements by Western blot orimmunohistochemistry of P70S6K, 4E-BP1, andAkt phos-phorylation. Because thesemarkersmay lack the requiredselectivity and sensitivity, there is a clear need for theidentification and validation of additional biomarkersallowing us to predict and monitor responses to mTORinhibitors. We propose p-mTOR S2481 as the more precisemarker to monitor response to mTOR kinase inhibition inmultiple myeloma cells, specifically when it involvesevaluating mTORC2/Akt function.

    Reportedly, mTOR is phosphorylated differentiallywhen associated with mTORC1 andmTORC2. Specifical-ly, mTOR phosphorylated on S2448 with Raptor, andmTOR phosphorylated on S2481 with Rictor. Therefore, itwas believed that mTORC1 contains mTOR phosphory-lated predominantly on S2448, whereas mTORC2 containsmTOR phosphorylated predominantly on S2481. More-over, intact mTORC2 is obligatory for mTOR S2481 phos-phorylation (13); thus, p-mTOR S2481 is a marker ofTORC2 activity (13, 14). In agreement with these data,our study in multiple myeloma cells found that baselineexpression of p-mTORS2481 correlatedwithTORC2-phos-phorylatable Akt (S473). AZD8055, unlike rapamycin, con-sistently inhibited mTOR phosphorylation at S2481 alongwith p-Akt S473. Importantly, p-mTOR S2481/p-Akt S473

    coexpression in multiple myeloma cell lines correlatedwith sensitivity to mTOR kinase inhibition. Our datatherefore suggest that phosphorylation of S2481 in mTORmay serve as a potential marker for intact mTORC2activity and sensitivity to mTOR kinase inhibitors inmultiple myeloma. However, further studies are war-ranted to confirm that p-mTOR S2481 is a fair readout ofmTORC2 activation in multiple myeloma.

    Until recently it was believed that TORC1/P70S6Ksuppression in the context of rapamycin treatment wasresponsible for triggering disinhibition of IGF1R signal-ing and subsequent Akt activation (20). In this regard,identification and characterization of mTORC2/Akt sig-naling and the discovery of mTOR kinase inhibitors

    Cirstea et al.

    Mol Cancer Ther; 13(11) November 2014 Molecular Cancer Therapeutics2498

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • demonstrating dual inhibition of TORC1 and TORC2pathways predicted the blockage of Akt signaling induc-ed in multiple myeloma cells by upstream IGF1R/PI3Kactivation through interaction with growth factors andthe BM microenvironment. In our study, AZD8055 inhi-bition of mTOR kinase in multiple myeloma cells asso-ciated with upregulation of the tyrosine phosphorylationsites (Y1135/1136) in the activation loop of the IGF1Rkinase domain. Furthermore, AZD8055-treated cells ex-posed to IGF1-sustained p-Akt S473 expression, whileTORC2 function remained inhibited, as suggested by in-hibition of p-mTOR S2481. Our observation aligned withrecent findings in other tumor types that Akt activationmay occur even in the context of mTOR kinase inhibitionbecause of blockage of TORC2/Akt and following dis-inhibition of RTK/PI3K feedback signaling stimulatedby growth factors (12, 19). Given that, it is possible thatAZD8055-triggered IGF1R activation is in part due tomTORC1 inhibition and in part secondary to TORC2/Aktsuppression.In multiple myeloma, activation of IGF1R induces sus-

    tained activation of PI3K/Akt and NF-kB, phosphoryla-tion of FKHR transcription factor, and upregulation of aseries of intracellular antiapoptotic proteins, includingFLIP, survivin, cIAP-2, and X-linked inhibitor of apopto-sis protein (XIAP), thereby decreasing drug sensitivity ofmultiple myeloma cells (21, 22). Moreover, IGF1R inhibi-tors have been shown to trigger significant multiple mye-loma cell toxicity (22, 23). A phase I study in multiplemyeloma demonstrated the safe profile of fugitumumab,the human monoclonal antibody directed against IGF1R,and some responses were reported in combination withdexamethasone (24). In our study, we observed IGF1/IGF1R-mediated reactivation of Akt and rescue of multi-ple myeloma cells from AZD8055-triggered apoptosis.Meanwhile, treatment with anti–IGF1R Ab enhancedAZD8055 activity and reversed IGF1-mediated rescue of

    multiple myeloma cells from AZD8055-induced cytotox-icity, suggesting that treatments targeting IGF1/IGF1Rsignaling are promising strategies and complementmTOR kinase inhibition in multiple myeloma.

    Our assertion that relief of feedback inhibition of IGF1Rin multiple myeloma tumors treated with mTOR kinaseinhibitors decreases the efficacy of PI3K pathway inhibi-tion has yet to be addressed in patients. We believe ourpreclinical findings will inform and complement theongoing clinical evaluation of mTOR kinase inhibition astargeted therapy and form the basis for testing them incombination with inhibitors of the IGF1R pathway.

    Disclosure of Potential Conflicts of InterestS.M.Guichard received other commercial research support fromAstra-

    Zeneca. No potential conflicts of interest were disclosed by the otherauthors.

    Authors' ContributionsConception and design: D. Cirstea, L. Santo, T. Hideshima, Y. Hu,F. Cottini, N. RajeDevelopment of methodology: N. Nemani, Y. Hu, F. Cottini, N. RajeAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): D. Cirstea, L. Santo, Y. Mishima, A. Mahindra,A. Yee, R. Suzuki, K.C. Anderson, N. RajeAnalysis and interpretation of data (e.g., statistical analysis, biostatis-tics, computational analysis): D. Cirstea, L. Santo, T. Hideshima, H. Eda,Y. Hu, H. Ohguchi, K.C. Anderson, N. RajeWriting, review, and/or revision of the manuscript: D. Cirstea, L. Santo,A. Mahindra, A. Yee, G. Gorgun, S.M. Guichard, K.C. Anderson, N. RajeAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): D. Cirstea, H. EdaStudy supervision: T. Hideshima, N. Raje

    Grant SupportN. Raje is a recipient of the Leukemia and Lymphoma Society clinical

    scholar Award.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.

    ReceivedApril 4, 2014; revisedAugust 8, 2014; acceptedAugust 16, 2014;published OnlineFirst August 29, 2014.

    References1. Palumbo A, Anderson K. Multiple myeloma. N Engl J Med 2011;

    364:1046–60.2. Munshi NC, Anderson KC. New strategies in the treatment of multiple

    myeloma. Clin Cancer Res 2013;19:3337–44.3. Laplante M, Sabatini DM. mTOR signaling in growth control and

    disease. Cell 2012;149:274–93.4. Watanabe R, Wei L, Huang J. mTOR signaling, function, novel inhibi-

    tors, and therapeutic targets. J Nucl Med 2011;52:497–500.5. Soliman GA, Acosta-Jaquez HA, Dunlop EA, Ekim B, Maj NE, Tee AR,

    et al. mTORSer-2481 autophosphorylationmonitors mTORC-specificcatalytic activity and clarifies rapamycin mechanism of action. J BiolChem 2010;285:7866–79.

    6. Sankhala K, Giles FJ. Potential of mTOR inhibitors as therapeuticagents in hematological malignancies. Expert Rev Hematol 2009;2:399–414.

    7. Raje N, Kumar S, Hideshima T, Ishitsuka K, Chauhan D, MitsiadesC, et al. Combination of the mTOR inhibitor rapamycin and CC-5013has synergistic activity in multiple myeloma. Blood 2004;104:4188–93.

    8. Cirstea D, Hideshima T, Rodig S, Santo L, Pozzi S, Vallet S, et al.Dual inhibition of akt/mammalian target of rapamycin pathway by

    nanoparticle albumin-bound-rapamycin and perifosine inducesantitumor activity in multiple myeloma. Mol Cancer Ther 2010;9:963–75.

    9. Wander SA, Hennessy BT, Slingerland JM. Next-generation mTORinhibitors in clinical oncology: how pathway complexity informs ther-apeutic strategy. J Clin Invest 2011;121:1231–41.

    10. Maiso P, Liu Y, Morgan B, Azab AK, Ren P, Martin MB, et al.Defining the role of TORC1/2 in multiple myeloma. Blood 2011;118:6860–70.

    11. Carew JS, Kelly KR, Nawrocki ST. Mechanisms of mTOR inhibitorresistance in cancer therapy. Target Oncol 2011;6:17–27.

    12. Rodrik-Outmezguine VS,Chandarlapaty S, PaganoNC,PoulikakosPI,Scaltriti M,Moskatel E, et al. mTOR kinase inhibition causes feedback-dependent biphasic regulation of AKT signaling. Cancer Discov2011;1:248–59.

    13. Copp J, Manning G, Hunter T. TORC-specific phosphorylation ofmammalian target of rapamycin (mTOR): phospho-Ser2481 is amarker for intact mTOR signaling complex 2. Cancer Res 2009;69:1821–7.

    14. OhWJ, Jacinto E.mTORcomplex 2 signaling and functions. Cell Cycle2011;10:2305–16.

    www.aacrjournals.org Mol Cancer Ther; 13(11) November 2014 2499

    Targeting mTOR Kinase/IGF1R Feedback in Multiple Myeloma

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • 15. Chresta CM, Davies BR, Hickson I, Harding T, Cosulich S, CritchlowSE, et al. AZD8055 is a potent, selective, and orally bioavailableATP-competitive mammalian target of rapamycin kinase inhibitorwith in vitro and in vivo antitumor activity. Cancer Res 2010;70:288–98.

    16. Tai YT, Teoh G, Shima Y, Chauhan D, Treon SP, Raje N, et al. Isolationand characterization of human multiple myeloma cell enriched popu-lations. J Immunol Methods 2000;235:11–9.

    17. Pirkmajer S, Chibalin AV. Serum starvation: caveat emptor. Am JPhysiol Cell Physiol 2011;301:C272–9.

    18. Qin L, Wang Y, Tao L, Wang Z. AKT down-regulates insulin-likegrowth factor-1 receptor as a negative feedback. J Biochem 2011;150:151–6.

    19. Chandarlapaty S, Sawai A, Scaltriti M, Rodrik-Outmezguine V, Grbo-vic-Huezo O, Serra V, et al. AKT inhibition relieves feedback suppres-sion of receptor tyrosine kinase expression and activity. Cancer Cell2011;19:58–71.

    20. Shi Y, Yan H, Frost P, Gera J, Lichtenstein A. Mammalian target ofrapamycin inhibitors activate the AKT kinase in multiple myeloma cells

    byup-regulating the insulin-likegrowth factor receptor/insulin receptorsubstrate-1/phosphatidylinositol 3-kinase cascade. Mol Cancer Ther2005;4:1533–40.

    21. Ge NL, Rudikoff S. Insulin-like growth factor I is a dual effector ofmultiple myeloma cell growth. Blood 2000;96:2856–61.

    22. Mitsiades CS, Mitsiades N, Poulaki V, Schlossman R, Akiyama M,Chauhan D, et al. Activation of NF-kappaB and upregulation of intracel-lular anti-apoptotic proteins via the IGF-1/Akt signaling inhumanmultiplemyeloma cells: therapeutic implications. Oncogene 2002;21:5673–83.

    23. WuKD, Zhou L, Burtrum D, Ludwig DL, Moore MA. Antibody targetingof the insulin-like growth factor I receptor enhances the anti-tumorresponse of multiple myeloma to chemotherapy through inhibition oftumor proliferation and angiogenesis. Cancer Immunol Immunother2007;56:343–57.

    24. LacyMQ,AlsinaM, FonsecaR,PaccagnellaML,MelvinCL,YinD, et al.Phase I, pharmacokinetic and pharmacodynamic study of the anti-insulinlike growth factor type 1 receptor monoclonal antibody CP-751,871 in patients with multiple myeloma. J Clin Oncol 2008;26:3196–203.

    Mol Cancer Ther; 13(11) November 2014 Molecular Cancer Therapeutics2500

    Cirstea et al.

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/

  • 2014;13:2489-2500. Published OnlineFirst August 29, 2014.Mol Cancer Ther Diana Cirstea, Loredana Santo, Teru Hideshima, et al. Inhibitor, AZD8055, in Multiple MyelomaDelineating the mTOR Kinase Pathway Using a Dual TORC1/2

    Updated version

    10.1158/1535-7163.MCT-14-0147doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://mct.aacrjournals.org/content/suppl/2014/08/29/1535-7163.MCT-14-0147.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://mct.aacrjournals.org/content/13/11/2489.full#ref-list-1

    This article cites 24 articles, 12 of which you can access for free at:

    Citing articles

    http://mct.aacrjournals.org/content/13/11/2489.full#related-urls

    This article has been cited by 2 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://mct.aacrjournals.org/content/13/11/2489To request permission to re-use all or part of this article, use this link

    on June 23, 2021. © 2014 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Published OnlineFirst August 29, 2014; DOI: 10.1158/1535-7163.MCT-14-0147

    http://mct.aacrjournals.org/lookup/doi/10.1158/1535-7163.MCT-14-0147http://mct.aacrjournals.org/content/suppl/2014/08/29/1535-7163.MCT-14-0147.DC1http://mct.aacrjournals.org/content/13/11/2489.full#ref-list-1http://mct.aacrjournals.org/content/13/11/2489.full#related-urlshttp://mct.aacrjournals.org/cgi/alertsmailto:[email protected]://mct.aacrjournals.org/content/13/11/2489http://mct.aacrjournals.org/

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages false /GrayImageMinResolution 200 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages false /MonoImageMinResolution 600 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 900 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > /FormElements false /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles false /MarksOffset 18 /MarksWeight 0.250000 /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /NA /PageMarksFile /RomanDefault /PreserveEditing true /UntaggedCMYKHandling /LeaveUntagged /UntaggedRGBHandling /LeaveUntagged /UseDocumentBleed false >> > ]>> setdistillerparams> setpagedevice