28
1 Targeting ribonucleotide reductase M2 and NF-kB activation with Didox to circumvent tamoxifen resistance in breast cancer Khyati N. Shah 1* , Elizabeth A. Wilson 1* , Ritu Malla 1 , Howard L. Elford 2 and Jesika S. Faridi 1 1 Department of Physiology and Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton CA 95211 2 Molecules for Health, Inc., Richmond, VA 23219, USA. *K.N. Shah and E.A. Wilson are co–first authors who contributed equally to this article. Running Title: Didox circumvents tamoxifen resistance Keyords: RRM2, Didox, Tamoxifen Grant Support: This work was supported, in part, by a Graduate Student Research Grant (K.N. Shah), a SAAG intramural fellowship (J.S. Faridi), and the Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific. Corresponding Author: Jesika S. Faridi, PhD Thomas J. Long Long School of Pharmacy University of the Pacific 751 Brookside Road Stockton CA 95211 Phone: (209) 946-2964 Fax: (209) 946-2857 Email: [email protected] Disclosure of Potential Conflicts of Interest: Dr. Howard L. Elford is an employee and shareholder in Molecules for Health Inc. on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

Khyati N. Shah1*, Elizabeth A. Wilson1*, Ritu Malla , Howard L. Elford and Jesika … · 2015. 9. 2. · 1 Targeting ribonucleotide reductase M2 and NF-kB activation with Didox to

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • 1

    Targeting ribonucleotide reductase M2 and NF-kB activation with Didox to circumvent tamoxifen resistance in breast cancer

    Khyati N. Shah1*, Elizabeth A. Wilson1*, Ritu Malla1, Howard L. Elford2 and Jesika S. Faridi1

    1Department of Physiology and Pharmacology, Thomas J. Long School of Pharmacy & Health

    Sciences, University of the Pacific, Stockton CA 95211

    2 Molecules for Health, Inc., Richmond, VA 23219, USA.

    *K.N. Shah and E.A. Wilson are co–first authors who contributed equally to this article.

    Running Title: Didox circumvents tamoxifen resistance

    Keyords: RRM2, Didox, Tamoxifen

    Grant Support: This work was supported, in part, by a Graduate Student Research

    Grant (K.N. Shah), a SAAG intramural fellowship (J.S. Faridi), and the

    Thomas J. Long School of Pharmacy and Health Sciences, University of the

    Pacific.

    Corresponding Author: Jesika S. Faridi, PhD Thomas J. Long Long School of Pharmacy

    University of the Pacific

    751 Brookside Road

    Stockton CA 95211

    Phone: (209) 946-2964

    Fax: (209) 946-2857

    Email: [email protected]

    Disclosure of Potential Conflicts of Interest: Dr. Howard L. Elford is an employee and shareholder in Molecules for Health Inc.

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 2

    Abstract: Tamoxifen is widely used as an adjuvant therapy for patients with estrogen receptor (ER-α)

    positive tumors. However, the clinical benefit is often limited due to the emergence of drug

    resistance. In this study, overexpression of ribonucleotide reductase M2 (RRM2) in MCF-7 breast

    cancer cells resulted in a reduction in the effectiveness of tamoxifen, through downregulation of ER-

    α66 and upregulation of the 36 kDa variant of ER (ER-α36). We identified that NF-κB, HIF-1α, and

    MAPK/JNK are the major pathways that are affected by RRM2 overexpression and result in

    increased NF-κB activity and increased protein levels of EGFR, HER2, IKK’s, Bcl-2, RelB, and p50.

    RRM2 overexpressing cells also exhibited higher migratory and invasive properties. Through time-

    lapse microscopy and protein profiling studies of tamoxifen-treated MCF-7 and T-47D cells, we have

    identified that RRM2 along with other key proteins is altered during the emergence of acquired

    tamoxifen resistance. Inhibition of RRM2 using siRRM2 or the ribonucleotide reductase (RR)

    inhibitor Didox not only eradicated and effectively prevented the emergence of tamoxifen resistant

    populations, but also led to the reversal of many of the proteins altered during the process of acquired

    tamoxifen resistance. Since Didox also appears to be a potent inhibitor of NF-κB activation,

    combining Didox with tamoxifen treatment cooperatively reverses ER-α alterations and inhibits NF-

    κB activation. Finally, inhibition of RRM2 by Didox reversed tamoxifen-resistant in vivo tumor

    growth and decreased in vitro migratory and invasive properties, revealing a beneficial effect of

    combination therapy that includes RRM2 inhibition to delay or abrogate tamoxifen resistance.

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 3

    Introduction: Estrogen receptor-α (ER-α) plays a fundamental role in the etiology and the progression of

    human breast cancer (1). Many therapies have been designed to inhibit the tumor-promoting effects

    of ER-α; however, tamoxifen has been the drug of choice for all stages of ER-positive breast cancer.

    As adjuvant therapy, tamoxifen reduces the risk of recurrence and improves overall survival (OS) in

    early breast cancer patients (2). Tamoxifen arrests cells in G0/G1 and decreases expression of several

    ERα target genes (3). Apart from blocking classical ER action, tamoxifen also induces DNA damage

    and apoptosis in ER-positive cells (4). Despite these benefits, some tumors recur due to acquired

    tamoxifen resistance giving rise to a sub-population of unresponsive cells (5). Several mechanisms of

    acquired tamoxifen resistance have been reported including down-regulation of ER-α expression (6).

    Tamoxifen resistance has also been linked to crosstalk between ER-α and signaling pathways

    involving Epidermal Growth Factor receptor (EGFR), HER2/ERBB2, or insulin-like growth factor

    receptor-I (IGFI-R) (7).

    ER-α66 is the classical ER-α of 66-kDa and is often referred to as simply “ER”. Cells which

    have high levels of ER-α66 are often termed ER-positive, while those lacking ER-α66 are called ER-

    negative. Clinical evidence suggests that approximately 40% of ER-α66 positive breast cancers also

    express a 36-kDa variant of ER-α (ER-α36), and this subset of patients is less likely to benefit from

    tamoxifen treatment (8, 9). Alternatively, endocrine resistant cells can develop a compensatory

    signaling pathway downstream of ER which results in hyperproliferation and increased cancer cell

    survival. In this type of resistance, ER-α36 may promote downstream signaling, such as the

    phosphatidylinositol 3-kinase (PI3K) pathway (10). We have previously shown that breast cancer

    cells overexpressing activated AKT exhibit tamoxifen-stimulated cell proliferation and enhanced cell

    motility. Moreover, we identified that RRM2 was a key contributor to AKT-induced tamoxifen

    resistance (11).

    Tamoxifen chemotherapy initially arrests tumor growth, but upon acquiring resistance, DNA

    synthesis is reactivated. The first step in DNA synthesis is conversion of ribonucleotides to their

    corresponding deoxyribonucleotides, catalyzed by the enzyme ribonucleotide reductase (RR) (12).

    This reaction is also the rate limiting step in DNA synthesis and cell division, and its activity is

    closely correlated with tumor growth rate and cell division (12). RR is composed of RRM1 and

    RRM2. Although the levels of the RRM1 protein does not change substantially during the cell cycle,

    there is an S-phase correlated increase in the RRM2 protein (13). The activity of RR, and therefore

    DNA synthesis and cell proliferation, are controlled by RRM2. In contrast, p53R2 (RRM2B) is

    involved in supplying dNTPs for DNA repair during mitochondrial DNA synthesis in the G0/G1

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 4

    phase of the cell cycle (14). Overexpression of the RRM2 subunit has been associated with malignant

    transformation and confers gemcitabine resistance (15). RRM2 functions in coordination with the S

    phase checkpoint to regulate DNA damage, replication stress, and genomic instability including

    mutagenesis (16, 17). Since chronic incubation of tamoxifen induces DNA damage, cells may

    upregulate RRM2 in an attempt to repair the DNA damage and are thus able to survive as a resistant

    population.

    Due to the DNA-damaging property of tamoxifen, combination therapies that enhance

    tamoxifen activity are of clinical benefit. RRM2 is an attractive target for combination therapy as it is

    overexpressed in breast cancer and contributes to development of resistance (11). Didox (3, 4-

    dihydroxybenzohydroxamic acid), is a strong inhibitor of RR that interferes with DNA synthesis and

    repair by blocking the production of deoxyribonucleotides and has demonstrated anti-tumor effects

    for decades (12, 18-20). Phase I/II clinical trial studies in cancer patients showed minimal toxicity

    and determined that the maximum tolerated dose of Didox is 6 g/m2, yielding peak plasma levels of

    425 μM (21-22). Didox can be tolerated by cancer patients at high dosages without major side

    effects, making Didox attractive for use in clinical applications. We previously reported that the

    combination of Didox and tamoxifen significantly reduced cell proliferation in AKT-induced

    tamoxifen resistance (11).

    Here, using gain and loss of RRM2, we show that RRM2 is sufficient to confer tamoxifen

    resistance in breast cancer cells. We demonstrate that RRM2 is associated with increased NF-κB

    activity, ER-α alterations, HER2 and EGFR upregulation, and increases in anti-apoptotic pathways.

    Finally, Didox significantly inhibits tamoxifen induced in vivo tumor growth. Our results show that

    Didox works synergistically with tamoxifen for the treatment and prevention of resistant breast

    cancer cells. Our data provide a preclinical rationale for evaluating tamoxifen in combination with

    Didox for breast cancer treatment.

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 5

    Materials and methods Cell culture and treatment

    MCF-7, T47D, HCC1428, BT483, ZR-75-30, ZR-75-1, SKBR3, BT20, BT549,

    HCC2157, MDA-MB-468, and MDA-MB-231 cells were purchased from ATCC between July

    and December 2012. Revived cells were used within 15 passages and cultured for less than 6

    months. Cells were maintained in DMEM/F12 supplemented with 10% FBS, streptomycin, and

    penicillin (Life Technologies). Didox (D) was supplied by Molecules for Health. For experiments,

    cells were treated with phenol-red-free, serum-free DMEM/F12 containing 0.1µM ethanol as vehicle

    (C), 0.1µM 17β-estradiol (E, Sigma), 1µM 4-hydroxy-tamoxifen (T, Sigma), 30µM D or a

    combination of 1µM T + 30µM Didox (T+D) for 24 hours.

    Western blot analyses Western blotting was performed as previously described (11, 23). Briefly, cells were

    disrupted in RIPA buffer (Sigma) or tumors were homogenized in lysis buffer (Cell Signaling)

    supplemented with aprotinin, leupeptin, and okadaic acid (Sigma). Lysates were clarified by

    centrifugation, and equal protein (75μg) was used for Western blotting. RRM1, RRM2, RRM2B

    (Sigma), p53, ER-α66 (Santa Cruz), ER-α36 (Alpha Diagnostics), GAPDH, EGFR, HER2, Bcl-2,

    pAKT, pER (S167) pERK, total ERK, total γH2AX, IKK sampler kit, NF-κB sampler kit, PARP,

    Caspase-9 (Cell Signaling), and pγ-H2AX (Millipore) were used for immunoblotting with secondary

    antibodies conjugated with IRDye 800CW or 680RD (LI-COR Biosciences) and visualized with a

    LI-COR Odyssey Imager.

    Meta-analysis of Breast Cancer Datasets RRM2 expression profiles and clinico-pathologic data of breast cancer patients were obtained

    from publicly available breast cancer microarray (25-29) and NCBI GEO (30-34) datasets. Data

    from tamoxifen-treated ER-positive patients were classified as tamoxifen-resistant if metastasis was

    indicated. Oncomine was used for data collection and analyses. P

  • 6

    Generation of Stable RRM2 overexpressing MCF-7 breast cancer cell lines MCF-7 cells were transfected with pCMV6-RRM2-myc-DDK or vector (Origene) using

    Fugene HD (Roche) and grown under geneticin selection after 48 hours. Clones overexpressing

    RRM2 were expanded to generate stable expressing clones MCF-7/R2.1, MCF-7/R2.3 and

    population MCF-7/R2.pool. The population MCF-7-VC cells stably express vector. Stable cells were

    routinely tested and authenticated according to the ATCC guidelines.

    Transcription factor reporter assays MCF-7/R2.1 and MCF-7 VC cells were reverse transfected onto the Cignal Finder 10-

    Pathway Reporter Array or the Cignal NF-κB Luciferase Reporter using SureFECT according to

    manufacturer’s protocol (Qiagen). Cells were treated for 24 hours with C, T, D, or T+D, as

    indicated, in phenol-red-free, serum-free DMEM/F12. Relative transcriptional activity was measured

    using the Dual Luciferase Assay (Promega). Three independent transfections were performed.

    siRNA-mediated suppression of RRM2 siRNA oligos targeting RRM2 (siRRM2) were designed and synthesized at Genentech Inc.

    Nontargeting siRNA and Dharmafect-1 were purchased (Dharmacon). Cells were transfected with

    siRNA by reverse transfection according to the manufacturers' directions. Transfection efficiencies

    were evaluated relative to nontargeting control by RT-qPCR, and the suppression of RRM2

    expression was sustained through day 7 for all breast cancer cell lines tested.

    Pathway-specific expression arrays Total RNA was isolated using RNeasy according to the manufacturer’s protocol

    (Qiagen). All RNA samples were examined for their concentration, purity, and integrity. The human

    Breast Cancer PCR array and the ECM and Adhesion Molecule (SABioscience) was used to assess

    the expression of 84 breast cancer genes according to the manufacturer’s instructions. Data shown

    represent the average of two replicates and were normalized using the previously validated

    housekeeping gene RPL13A levels (23).

    Establishment and treatment of acquired tamoxifen resistant cells

    MCF7 and T-47D were treated with 1μM tamoxifen and dose was increased every 10 days

    until 5μM. Once established, TamR resistant cells were maintained in continuous culture with 1μM

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 7

    tamoxifen. For protein profiling, replicate plates were treated with tamoxifen in this manner and

    lysates were collected over 30 days. Control lysates were collected after 3 days of treatment with

    vehicle. TamR lysates were collected with continuous treatment with 1μM tamoxifen alone or

    combined with Didox for five days.

    Cell proliferation and motility assays

    For proliferation, 1000 cells/well were plated in phenol-red-free, DMEM-F12 medium with

    2% CSS. After 24 hours, treatment media were replenished on alternate days. On assay days,

    CellTiter-96 Aqueous One Solution (Promega) was added, incubated for 1 hour, and measured at

    490nm. For the colony formation assay, 3000 cells/well were cultured in 5% FBS phenol red-free

    DMEM. The following day, the cells were treated and allowed to grow for 14 days. Colonies were

    stained with crystal violet and analyzed. A modified scratch assay was performed by plating 20,000

    cells in wells containing inserts (IbIDI Martinsried). After 24 hours inserts were removed, and the

    percent open area was calculated after 20 additional hours. Cell migration and invasion experiments

    were carried out using the QCM™ 24-well Colorimetric Cell Migration kit or the Invasion Assay kit

    (Chemicon) according to manufacturer’s protocols. These experiments were conducted in triplicate

    and data is shown as the mean ± SEM.

    Synergy determinations The IC50 value of tamoxifen and Didox was determined for MCF-7 VC, MCF-7TamR and

    RRM2 overexpressing MCF-7 cells (MCF-7/R2.1). Based on the IC50 value, fixed dose ratios were

    used to determine five different drug combinations (i.e. 2X, 1X, X, X/2, X/4, where X: IC50 of an

    individual drug). Synergistic, additive, or antagonistic effects were determined using the combination

    index (CI) method developed by Chou and Talalay. Synergy, additivity, and antagonism are

    indicated by CI values of 1, respectively (24).

    Xenograft studies All animal procedures were approved by the Institutional Animal Care and Use Committee of

    the University of the Pacific. RRM2 overexpressing MCF-7 cells (4 x 106/site) were subcutaneously

    injected into the flank of ncr nude female ovariectomized nude mice (6-15 tumors/group). Mice

    were implanted with estrogen (0.72 mg/pellet, Innovative Research of America, (IRA)) and

    tamoxifen as indicated (5 mg/pellet, IRA). Beginning day 7 as indicated, daily Didox (N, 3,4-

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 8

    Dihydroxybenzohydroxamic, 200 or 425 mg/kg/day) was injected intraperitoneally after compound

    was freshly dissolved in water and filtered. The in vivo tumor volume was approximated using the

    formula for an ellipsoid (4/3π(r1)2(r2)) where r1

  • 9

    Results: RRM2 expression inversely correlates with ER expression in breast cancer RRM2 expression is upregulated in cancer and is associated with increased tumor

    aggressiveness, poor prognosis, and chemoresistance (11, 14-16). Here, we demonstrate that RRM2

    expression is significantly upregulated in ER-negative as compared to ER-positive breast cancer

    cells, as is a 36kDa variant of ER (ER-α36) (Fig. 1A). Interestingly, of the ER-positive cells, ZR-75-

    1 alone has high levels of RRM2 and ER-α36. We have previously shown that ZR-75-1 cells exhibit

    tamoxifen-resistant cell proliferation and that inhibition of RRM2 using siRRM2 not only restores

    tamoxifen sensitivity, but it also represses the DNA repair enzymes that protect these cells from

    tamoxifen-induced apoptosis (11).

    Meta-analysis of five different Oncomine datasets revealed that RRM2 is highly expressed in

    ER-negative tumors (Fig. 1B, Supplementary Table S1A) (25-29). Using survival data from

    tamoxifen-treated ER-positive patients, RRM2 mRNA expression was determined to be significantly

    higher in patients who experienced tumor relapse (Fig. 1C, Supplementary Table S1B) (30-34).

    Kaplan-Meier survival plots (KMplots) of tamoxifen-treated ER-positive patients indicate that

    increased RRM2 expression is strongly correlated with reduced RFS and OS (Fig. 1D, 1E) (35).

    Stable overexpression of RRM2 reduces tamoxifen sensitivity in MCF-7 cells We previously reported that inhibition of RRM2 reverses tamoxifen resistance in breast

    cancer cells (11). To strengthen the association between RRM2 and tamoxifen resistance, we

    transiently overexpressed RRM2 in ER-positive, tamoxifen-sensitive MCF-7 and T-47D cells and

    confirmed RRM2 expression by Western blot analysis (Supplementary Fig. S1). Relative to vector

    control cells (VC), RRM2 overexpressing MCF-7 and T-47D cells exhibit reduced tamoxifen

    sensitivity and higher IC50 values resulting in resistance ratios of 3.2-3.3. Stable RRM2

    overexpressing (clones MCF-7/R2.1, MCF/R2.2, and pool population MCF-7/R2.Pool) and vector

    control (MCF-7 VC) MCF-7 cells were then generated (Supplementary Fig. S2). As expected, MCF-

    7 VC cell proliferation was significantly inhibited with 1µM tamoxifen. In contrast, RRM2

    overexpressing MCF-7 cells demonstrated a loss of sensitivity to tamoxifen. Treatment with the RR

    inhibitor Didox inhibited the cell proliferation of MCF-7/R2 cells when given alone and was even

    more potent when given in combination with tamoxifen. Interestingly, RRM2 overexpressing cells

    had a slightly diminished estrogen response as compared to MCF-7 VC cells. Since all three RRM2

    overexpressing stable lines had similar properties, we used MCF-7/R2.1 cells for subsequent

    experiments.

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 10

    RRM2 overexpression increases NF-κB activity and gene transcription in breast cancer cells RRM2 overexpression specifically upregulates the NF-κB, HIF1-α, and MAPK/JNK

    proliferation and differentiation pathways (Fig. 2A). Interestingly, Didox significantly inhibited NF-

    κB (p

  • 11

    S536 indicating increased NF-κB activity. When examining apoptotic proteins, we observed an

    initial spike (days 1- 5) then decrease (day 10) in cleaved PARP and Caspase-9 leading to the

    increase in the anti-apoptotic factor Bcl-2 (Fig. 3D). Finally, the emergence of tamoxifen resistant

    populations occurred around day 15 which coincides with the activation of pERK1/2 and pAKT

    (S473) kinases.

    Combining tamoxifen and Didox circumvents resistance via cooperatively reducing ER alterations and NF-κB signaling

    To evaluate the efficacy of tamoxifen and Didox (T+D) combination therapy on the

    emergence of tamoxifen resistance, time-lapsed images were captured and significant cell rounding

    (indicative of cell death) was observed as early as day 1. Treatment with T+D prevented the

    emergence of tamoxifen resistance in parental cells and successfully eradicated TamR lines (Fig. 4A,

    4B). Didox reduced RRM2, ER-α36 and S167 phosphorylation of ERα, and simultaneously

    increased ER-α66 levels (Fig. 4C, 4D). Moreover, combination therapy induced apoptosis as

    indicated by increased S139 phosphorylation of γ-H2AX, increased cleavage of PARP and Caspase-

    9, reduced levels of the anti-apoptotic Bcl-2, resensitizing RRM2 overexpressing and TamR cells to

    tamoxifen-induced cell death (Supplementary Fig. S3). Furthermore, Didox reduced the expression

    of EGFR, HER2, pERK, and pAKT in T-47DTamR (Fig. 4D), and all but pAKT in MCF-7TamR

    cells (Fig. 4B).

    Since there was an observed increase in NF-κB signaling in TamR cells, we sought to

    examine the effect of T+D in this pathway. Didox reduces expression of the NF-κB related IKK’s,

    p50, RelB, pIKK, p-IκBα, and the S536 phosphorylation of p65 (Fig. 4B, 4D). Using an NF-κB

    reporter assay, we found that Didox alone, and more significantly, T+D downregulates NF-κB

    activity in MCF-7/R2.1, MCF-7TamR, and T-47DTamR cells (Fig. 4E).

    Didox cooperates with tamoxifen to reduce cell proliferation and tumor growth To evaluate the combined effects of T+D, cellular viability was evaluated using the

    combination index (CI) (24). Compared to single-agent treatments, the combination of T+D

    significantly reduced cell viability in MCF-7/R2.1 and MCF-7TamR cells (Supplementary Fig. S4).

    Combination index values of 0.75 and 0.47 indicate synergistic actions of 1.25µM tamoxifen and

    37.5µM Didox in MCF-7/R2.1 and MCF-7TamR (but not parental MCF-7) cells respectively on

    reducing cell proliferation. Cell viability and toxicity studies indicate that with tamoxifen treatment,

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 12

    MCF7/R2.1 and MCF7TamR cells exhibit higher viability, lower toxicity, and higher IC50 than

    parental MCF-7 cells (Supplementary Fig. S5).

    To determine the in vivo efficacy tamoxifen and Didox (T+D) combination therapy in RRM2

    overexpressing breast tumors, MCF-7/R2.1 cells were subcutaneously injected in mice treated with

    tamoxifen as indicated. Daily Didox (200 or 425 mg/kg/day) treatment was begun as indicated seven

    days after cell injection. Mice treated with tamoxifen exhibited significantly greater tumor growth as

    compared to mice treated with the combination of T+D200 (p

  • 13

    Discussion: Tamoxifen effectively blocks estrogen stimulated tumor growth by inhibiting the activity of

    ER-α66 in breast cancer cells (2). Overexpression of tyrosine kinase signaling pathways and

    subsequent downregulation of ER-α66 after long term tamoxifen treatment is responsible for the

    development of acquired tamoxifen resistance in ER-α66-positive primary tumors (3, 5-8). As well,

    tamoxifen resistance can contribute to greater breast tumor aggressiveness (37). Although the

    mechanisms underlying acquired tamoxifen resistance are largely unknown, we have demonstrated

    that RRM2 is upregulated in AKT-induced tamoxifen resistant breast cancer cells and that inhibition

    of RRM2 by siRNA significantly overturns this resistance (11). This current study shows that RRM2

    overexpression alone is sufficient to promote tamoxifen resistance, is expressed during the

    emergence of de novo tamoxifen resistance (Fig. 3C), and downregulates ESR1 (ER-α) gene and

    protein expression (Fig. 2B, 3C). Using in vitro breast cancer cells, patient datasets, tissue

    microarrays, and tumor xenografts, we report for the first time that there is an inverse correlation

    between RRM2 and ER-α66 and a direct correlation with ER-α36. By inhibiting RRM2 using

    Didox, we show that the emergence of acquired tamoxifen resistance is circumvented, while the

    downregulation of ER-α66 and upregulation of ER-α36 is reversed.

    Our data suggest that ER-α66 downregulation in RRM2 overexpressing breast cancers may

    occur through increases in NF-κB activation and signaling. Transrepression of ER by NF-κB has

    been proposed as a mechanism by which ER-positive breast tumor cells lose ER expression and,

    hence, give rise to a subpopulation of tumor cells that are resistant to endocrine treatment (38, 39).

    Furthermore, we provide evidence that increased NF-κB signaling leading to ERα alterations are

    mediated via RRM2 overexpression and can be reversed using Didox through its ability to inhibit

    NF-κB activation (Fig. 4E). We have shown that the inhibition of RRM2 by Didox was able to

    eradicate MCF-7TamR and T-47DTamR populations by day 15, supporting the use of Didox to

    resensitize breast cancer cells to tamoxifen therapy. More importantly, Didox prevented the

    emergence of tamoxifen resistance in two models, suggesting the therapeutic use of Didox to also

    prevent the emergence of tamoxifen resistance (Fig. 4A, 4B).

    Here, by overexpressing RRM2, we were able to reproduce the tamoxifen-resistant

    phenotype and have identified that NF-κB, HIF-1α, and MAPK/JNK are the major pathways that are

    affected (Fig.2A). These pathways have been shown to play a significant role in mammary-

    carcinogenesis and have been implicated in tumor resistance. In particular, the NF-κB pathway has

    been shown to regulate cell proliferation, differentiation, and invasion (39). Similar to our study, the

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 14

    NF-κB pathway was reported to be increased by RRM2 and inhibited by Didox, strengthening the

    findings that RRM2 is a key regulator of the NF-κB pathway (40, 41). Although Didox exerts its

    activity by destabilizing RR through its free radical scavenging and iron chelating properties, Didox

    has also been shown to have strongly inhibit NF-κB activation (42).

    Due to tamoxifen’s ability to induce cell death by causing DNA damage, we sought to

    determine whether RRM2 plays in role in protecting breast cancer cells from tamoxifen-induced cell

    death (4). It is likely that cells having high RRM2 (cells in S-phase or in drug induced upregulation

    of RRM2) in a heterogeneous population survive and emerge as a resistant population that is

    protected from cell death due to DNA damage. Others have shown that RRM2 regulates anti-

    apoptotic proteins such as Bcl-2 in certain cancers (43). Here, we show that RRM2 overexpression

    protects against tamoxifen-induced apoptosis and results in lowered S139 γH2AX phosphorylation,

    reduced PARP and Caspase-9 cleavage, and increased Bcl-2 levels, which are well-established

    mechanisms of chemoresistance (44). These RRM2-induced protective effects were reversed by the

    combination of tamoxifen and Didox treatment (Fig. 2D). This is the first report that Didox can

    resensitize breast cancer cells to tamoxifen-induced cell death. Since high RRM2 expression is

    correlated with poor RFS and OS in tamoxifen-treated patients (Fig. 1D and 1E), RR inhibitors used

    in combination with tamoxifen may improve RFS and OS.

    Recently, there has been interest in drug combinations with greater efficacy and reduced

    toxicity. To date, no serious side effects have been reported with Didox treatment even when

    administered for long periods (21). Didox has been shown to inhibit proliferation of several cancer

    cell types (18-20, 45) and modulate other cancer chemotherapeutic agents resulting in elevated

    apoptosis (42). Didox was also shown to synergize with temozolomide in brain tumors and with

    doxorubicin in liver cancer cells (46, 47). Our study indicates that combining Didox with tamoxifen

    synergistically reduced cell in RRM2 overexpressing MCF-7, MCF-7TamR, and T-47DTamR cells

    (Fig. 5, Supplementary Fig. S4). Additionally, our study reports that Didox inhibits RRM2 induced

    cell motility, migration, and invasion (Fig. 6). Our data suggest that combining Didox with tamoxifen

    may offer significant benefits to patients with ER-positive breast cancer with high RRM2 expression

    or patients who have relapsed after long-term tamoxifen treatment. Similarly, RRM2 was recently

    suggested as a prognostic marker associated with poor survival and tamoxifen resistance, which

    supports our findings that RRM2 is an important contributor on the pathway to tamoxifen resistance

    (48).

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 15

    In summary, our findings strongly complement the current knowledge regarding the

    molecular mechanisms underlying acquired tamoxifen resistance in breast cancer and also provide

    strong evidence that RRM2 is important in regulating ER-α expression, hence responsiveness to

    tamoxifen. Our data demonstrate for the first time that overexpression of RRM2 in MCF-7 breast

    cancer cells leads to upregulation of EGFR and NF-κB signaling, downregulation of ER-α66

    expression, and upregulation of ER-α36, contributing to the generation of acquired tamoxifen

    resistance. Overexpression of RRM2 also enhances the proliferative capacity and the migratory and

    invasive ability of MCF-7 breast cancer cells. Furthermore, co-treatment of tamoxifen and Didox

    resulted in reduced proliferation rates with decreased in vitro migratory and invasive properties.

    Most importantly, the combination treatment produced significant tumor inhibition, suggesting a

    critical role of RRM2 in maintaining a malignant phenotype in breast cancer. These findings indicate

    that RRM2 may be potentially used as a prognostic factor in breast cancer patients undergoing

    tamoxifen therapy and can be considered a potential therapeutic target in tumors that have acquired

    resistance to tamoxifen. Further study of the molecular mechanisms by which RRM2 is activated

    during the development of acquired tamoxifen resistance in breast cancer will provide more detailed

    insights regarding the biological function of RRM2. Lastly, these data provide a rationale for the

    combination of tamoxifen and Didox therapy to circumvent or prevent tamoxifen resistance in breast

    cancer.

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 16

    List of Abbreviations Abbreviations

    ER-α, Estrogen receptor-α; C, Vehicle control; E, Estrogen; T, Tamoxifen; Tam,

    Tamoxifen; D, Didox; HBSS, Hank’s buffered salt solution; FBS, Fetal bovine serum; CSS,

    Charcoal-stripped-serum; RT-qPCR, Reverse Transcription Quantitative PCR Polymerase Chain

    Reaction; SEM, standard error of the mean; OS, overall survival; RFS, relapse free survival; DMFS,

    Distant Metastasis Free Survival; RR, ribonucleotide reductase; RRM2, ribonucleotide reductase

    M2; KMplots, Kaplan-Meier survival plots; SEM, standard error of the mean; SD, standard

    deviation. Acknowledgements

    The authors thank Dr. Arturo Cardounel and Dr. Murugesan Velayutham (University of

    Pittsburgh) for their clinical expertise and assistance with the tissue microarrays. The authors thank

    Mr. Mike Manzer (UC Davis, VMTH) for his immunohistological and digital imaging assistance.

    Finally, the authors thank Dr. John Livesey and Dr. Timothy Smith (Department of Physiology and

    Pharmacology, University of the Pacific, Thomas J. Long School of Pharmacy and Health

    Sciences) and Dr. Lisa Wrischnik (Department of Biological Sciences) for critical reading and

    constructive comments during the preparation of this manuscript.

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 17

    References: 1. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:10869-74. 2. Doisneau-Sixou SF, Sergio CM, Carroll JS, Hui R, Musgrove EA, Sutherland RL. Estrogen and antiestrogen regulation of cell cycle progression in breast cancer cells. Endocrine-related cancer. 2003;10:179-86. 3. Musgrove EA, Hamilton JA, Lee CS, Sweeney KJ, Watts CK, Sutherland RL. Growth factor, steroid, and steroid antagonist regulation of cyclin gene expression associated with changes in T-47D human breast cancer cell cycle progression. Molecular and cellular biology. 1993;13:3577-87. 4. Wozniak K, Kolacinska A, Blasinska-Morawiec M, Morawiec-Bajda A, Morawiec Z, Zadrozny M, et al. The DNA-damaging potential of tamoxifen in breast cancer and normal cells. Archives of Toxicology. 2007;81:519-27. 5. Arpino G, De Angelis C, Giuliano M, Giordano A, Falato C, De Laurentiis M, et al. Molecular mechanism and clinical implications of endocrine therapy resistance in breast cancer. Oncology. 2009;77 Suppl 1:23-37. 6. Ring A, Dowsett M. Mechanisms of tamoxifen resistance. Endocrine-related cancer. 2004;11:643-58. 7. Gee JM, Robertson JF, Gutteridge E, Ellis IO, Pinder SE, Rubini M, et al. Epidermal growth factor receptor/HER2/insulin-like growth factor receptor signalling and oestrogen receptor activity in clinical breast cancer. Endocrine-related cancer. 2005;12 Suppl 1:S99-S111. 8. Wang Z, Zhang X, Shen P, Loggie BW, Chang Y, Deuel TF. Identification, cloning, and expression of human estrogen receptor-alpha36, a novel variant of human estrogen receptor-alpha66. Biochemical and biophysical research communications. 2005;336:1023-7. 9. Li G, Zhang J, Jin K, He K, Zheng Y, Xu X, et al. Estrogen receptor-alpha36 is involved in development of acquired tamoxifen resistance via regulating the growth status switch in breast cancer cells. Molecular oncology. 2013;7:611-24. 10. Clark AS, West K, Streicher S, Dennis PA. Constitutive and inducible Akt activity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast cancer cells. Molecular cancer therapeutics. 2002;1:707-17. 11. Shah KN, Mehta KR, Peterson D, Evangelista M, Livesey JC, Faridi JS. AKT-induced tamoxifen resistance is overturned by RRM2 inhibition. Molecular cancer research : MCR. 2014;12:394-407. 12. Elford HL, Freese M, Passamani E, Morris HP. Ribonucleotide reductase and cell proliferation. I. Variations of ribonucleotide reductase activity with tumor growth rate in a series of rat hepatomas. Journal of Biological Chemistry Biol Chem. 1970; 245:5228-33. 13. Engstrom Y, Eriksson S, Jildevik I, Skog S, Thelander L, Tribukait B. Cell cycle-dependent expression of mammalian ribonucleotide reductase. Differential regulation of the two subunits. The Journal of biological chemistry. 1985;260:9114-6. 14. Pontarin G, Ferraro P, Bee L, Reichard P, Bianchi V. Mammalian ribonucleotide reductase subunit p53R2 is required for mitochondrial DNA replication and DNA repair in quiescent cells. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:13302-7. 15. Duxbury MS, Ito H, Zinner MJ, Ashley SW, Whang EE. Inhibition of SRC tyrosine kinase impairs inherent and acquired gemcitabine resistance in human pancreatic adenocarcinoma cells. Clinical cancer research : an official journal of the American Association for Cancer Research. 2004;10:2307-18.

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 18

    16. Aird KM, Zhang G, Li H, Tu Z, Bitler BG, Garipov A, et al. Suppression of nucleotide metabolism underlies the establishment and maintenance of oncogene-induced senescence. Cell reports. 2013;3:1252-65. 17. D'Angiolella V, Donato V, Forrester FM, Jeong YT, Pellacani C, Kudo Y, et al. Cyclin F-mediated degradation of ribonucleotide reductase M2 controls genome integrity and DNA repair. Cell. 2012;149:1023-34. 18. Elford HL, Wampler GL, van't Riet B. New ribonucleotide reductase inhibitors with antineoplastic activity. Cancer research. 1979;39:844-51. 19. Elford HL, Van't Riet B, Wampler GL, Lin AL, Elford RM. Regulation of ribonucleotide reductase in mammalian cells by chemotherapeutic agents. Advances in Enzyme Regulation. 1980;19:151-68. 20. van't Riet B, Wampler GL, Elford HL. Synthesis of hydroxy- and amino-substituted benzohydroxamic acids: inhibition of ribonucleotide reductase and antitumor activity. Journal of medicinal chemistry. 1979;22:589-92. 21. Veale D, Carmichael J, Cantwell BM, Elford HL, Blackie R, Kerr DJ, Kaye SB, Harris AL. A phase 1 and pharmacokinetic study of didox: a ribonucleotide reductase inhibitor. Br J Cancer. 1988 Jul;58(1):70-2. 22. Carmichael J, Cantwell BM, Mannix KA, Veale D, Elford HL, Blackie R, et al. A phase I and pharmacokinetic study of didox administered by 36 hour infusion. The Cancer Research Campaign Phase I/II Clinical Trials Committee. Br J Cancer. 1990;61:447-50. 23. Shah KN, Faridi JS. Estrogen, tamoxifen, and Akt modulate expression of putative housekeeping genes in breast cancer cells. Journal of Steroid Biochemistry and Molecular Biology. 2011;125:219-25. 24. Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacological reviews. 2006;58:621-81. 25. Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346-52. 26. Desmedt C, Piette F, Loi S, Wang Y, Lallemand F, Haibe-Kains B, et al. Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clinical cancer research : an official journal of the American Association for Cancer Research. 2007;13:3207-14. 27. Boersma BJ, Reimers M, Yi M, Ludwig JA, Luke BT, Stephens RM, et al. A stromal gene signature associated with inflammatory breast cancer. International journal of cancerJournal international du cancer. 2008;122:1324-32. 28. Lu X, Lu X, Wang ZC, Iglehart JD, Zhang X, Richardson AL. Predicting features of breast cancer with gene expression patterns. Breast cancer research and treatment. 2008;108:191-201. 29. Ma XJ, Dahiya S, Richardson E, Erlander M, Sgroi DC. Gene expression profiling of the tumor microenvironment during breast cancer progression. Breast cancer research : BCR. 2009;11:R7. 30. Zhang Y, Sieuwerts AM, McGreevy M, Casey G, Cufer T, Paradiso A, et al. The 76-gene signature defines high-risk patients that benefit from adjuvant tamoxifen therapy. Breast cancer research and treatment. 2009;116:303-9. 31. Loi S, Haibe-Kains B, Desmedt C, Lallemand F, Tutt AM, Gillet C, et al. Definition of clinically distinct molecular subtypes in estrogen receptor-positive breast carcinomas through genomic grade. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007;25:1239-46.

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 19

    32. Loi S, Haibe-Kains B, Desmedt C, Wirapati P, Lallemand F, Tutt AM, et al. Predicting prognosis using molecular profiling in estrogen receptor-positive breast cancer treated with tamoxifen. BMC genomics. 2008;9:239-2164-9-239. 33. Chanrion M, Negre V, Fontaine H, Salvetat N, Bibeau F, Mac Grogan G, et al. A gene expression signature that can predict the recurrence of tamoxifen-treated primary breast cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2008;14:1744-52. 34. Pawitan Y, Bjohle J, Amler L, Borg AL, Egyhazi S, Hall P, et al. Gene expression profiling spares early breast cancer patients from adjuvant therapy: derived and validated in two population-based cohorts. Breast cancer research : BCR. 2005;7:R953-64. 35. Gyorffy B, Lanczky A, Szallasi Z. Implementing an online tool for genome-wide validation of survival-associated biomarkers in ovarian-cancer using microarray data from 1287 patients. Endocrine-related cancer. 2012;19:197-208. 36. Sharma A, Singh K, Almasan A. Histone H2AX phosphorylation: a marker for DNA damage. Methods in molecular biology (Clifton, NJ). 2012;920:613-26. 37. Hiscox S, Jiang WG, Obermeier K, Taylor K, Morgan L, Burmi R, Barrow D, Nicholson RI. Tamoxifen resistance in MCF7 cells promotes EMT-like behaviour and involves modulation of beta-catenin phosphorylation. Int J Cancer 2006;118:290–301 38. Wang X, Belguise K, O'Neill CF, Sánchez-Morgan N, Romagnoli M, Eddy SF, et al. RelB NF-kappaB represses estrogen receptor alpha expression via induction of the zinc finger protein Blimp1. Mol Cell Biol. 2009;29:3832-44. 39. Sas L, Lardon F, Vermeulen PB, Hauspy J, Van Dam P, Pauwels P, et al. The interaction between ER and NFκB in resistance to endocrine therapy. Breast Cancer Res. 2012;14:212. 40. Duxbury MS, Whang EE. RRM2 induces NF-kappaB-dependent MMP-9 activation and enhances cellular invasiveness. Biochemical and biophysical research communications. 2007;354:190-6. 41. Lee R, Beauparlant P, Elford H, Ponka P, Hiscott J. Selective inhibition of l kappaB alpha phosphorylation and HIV-1 LTR-directed gene expression by novel antioxidant compounds. Virology. 1997 Aug 4;234:277-90. 42. Inayat MS, Chendil D, Mohiuddin M, Elford HL, Gallicchio VS, Ahmed MM. Didox (a novel ribonucleotide reductase inhibitor) overcomes Bcl-2 mediated radiation resistance in prostate cancer cell line PC-3. Cancer biology & therapy. 2002;1:539-45. 43. Kang MH, Reynolds CP. Bcl-2 inhibitors: targeting mitochondrial apoptotic pathways in cancer therapy. Clin Cancer Res. 2009;15:1126-32. 44. Elford HL, van't Riet B. Inhibition of nucleoside diphosphate reductase by hydroxybenzohydroxamic acid derivatives. Pharmacology & therapeutics. 1985;29:239-54. 45. Rahman MA, Amin ARMR, Wang D, Koenig L, Nannapaneni S, Chen Z, et al. RRM2 regulates Bcl-2 in head and neck and lung cancers: a potential target for cancer therapy. Clin Cancer Res. 2013;19:3416-28. 46. Figul M, Soling A, Dong HJ, Chou TC, Rainov NG. Combined effects of temozolomide and the ribonucleotide reductase inhibitors didox and trimidox in malignant brain tumor cells. Cancer chemotherapy and pharmacology. 2003;52:41-6. 47. Al-Abd AM, Al-Abbasi FA, Asaad GF, Abdel-Naim AB. Didox potentiates the cytotoxic profile of doxorubicin and protects from its cardiotoxicity. European journal of pharmacology. 2013;718:361-9. 48. Putluri N, Maity S, Kommangani R, Creighton CJ, Putluri V, Chen F, et al. Pathway-Centric Integrative Analysis Identifies RRM2 as a Prognostic Marker in Breast Cancer Associated with Poor Survival and Tamoxifen Resistance. Neoplasia. 2014;16:390-402.

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • 20

    Figure Legends

    Fig. 1: RRM2 inversely correlates with ER-α66 expression. (A) Western blot analysis of RRM2 and

    ER levels in ER-positive and ER-negative cells. (B) Meta-analysis demonstrates higher RRM2

    expression in ER-negative (dark box) as compared to ER-positive cancer patients (white box). (C)

    Tamoxifen-resistant patients (dark box) have higher RRM2 than tamoxifen-sensitive patients (white

    box. Fold change and p-values given in Supplementary Table S1. KMplots demonstrate

    overexpression of RRM2 is predictive of (D) lower RFS (p=4.9e-5) and (E) lower OS (p=0.00025) in

    tamoxifen-treated ER-positive patients.

    Fig. 2: RRM2 overexpression modulates NF-κB activity and gene transcription. (A) Reporter array shows NF-κB (p

  • 21

    mg/kg/day (D), n=6-15. Didox treatment was initiated at day 7 post-cell injection when RRM2

    overexpressing MCF-7 xenografts reach maximum tumor volumes (30 mm3) in the absence of

    tamoxifen. Mean tumor volume ± SEM is shown ** p

  • RRM2

    ER-α36

    ER-α66

    GAPDH

    ER-Positive ER-Negative

    A B

    -2

    0

    2

    4

    6

    8

    Re

    lati

    ve m

    RN

    A e

    xpre

    ssio

    n

    25 26 27 28 29

    ER-Positive ER-Negative

    C OS in Tam treated ER Positive patients D E

    -2

    0

    2

    4

    6

    8

    10

    Re

    lati

    ve m

    RN

    A e

    xpre

    ssio

    n

    30 31 32 33 34

    Tam-resistant Tam-sensitive

    Low RRM2

    High RRM2

    Pro

    bab

    ilit

    y o

    f R

    FS

    Low RRM2

    High RRM2

    Pro

    bab

    ilit

    y o

    f O

    S

    RFS in Tam treated ER positive patient

    Fig. 1

    HR=1.96 (1.41 – 2.73)

    P=4.9e-05

    HR=2.23 (1.43 – 3.46)

    P=0.00025

    Time (months) Time (months)

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • RRM2

    RRM1

    ER-α66

    ER-α36

    EGFR

    HER2

    GAPDH

    RRM2B

    Bcl-2

    p53

    IKK-δ

    IKK-β

    IKK-α

    IKK-ε

    IKK-γ

    p52

    p50

    p65

    RelB

    C-Rel

    PARP

    Cl-PARP

    Caspase9

    Cl-Caspase9

    C T D T+D C T D T+D

    MCF-7 MCF-7/R2.1

    pγ-H2AX (S139)

    γ-H2AX

    GAPDH

    -40

    -20

    0

    20

    40

    BC

    L2 IL6

    PA

    RP

    BIR

    C5

    CC

    ND

    2

    EGFR

    ERB

    B2

    MY

    C

    IGF1

    VEG

    FA

    MM

    P9

    CC

    ND

    1

    TWIS

    T1

    MG

    MT

    JUN

    EGF

    IGF1

    R

    AR

    CD

    H1

    TP5

    3

    AP

    C

    CD

    KN

    1A

    BA

    D

    ESR

    1

    CD

    KN

    2A

    Fold

    Ch

    ange

    -7 -4 -1 2 5 8

    NFKB

    HIF1-α

    MAPK/ERK

    MAPK/JNK

    Wnt

    Myc/max

    TGF-B

    pRB

    Notch

    p53

    Log2 (Fold Change)

    RRM2 +Didox

    RRM2 -Didox

    Increase Decrease

    * *

    *

    *

    *

    Fig. 2

    A B

    C

    D

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • ER-α66

    ER-α36

    ER-α66 (pS167)

    ER-β

    ESTROGEN RECEPTOR SIGNALING

    PANEL

    p52

    p50

    p65

    RelB

    p p65 (S53)

    GAPDH

    C-Rel

    NF-κB SIGNALING PANEL

    p53

    EGFR

    HER2

    pERK1/2

    Total ERK1/2

    pAKT (S473)

    Total AKT

    GROWTH RECEPTOR SIGNALING

    PANEL

    MCF-7 1 5 10 15 20 30 Days

    IKK-δ

    pIKK-α/β (S176/180)

    IKK-β

    IKK-α

    pIkBα (S32)

    Total IkBα

    IKK-ε

    IKK-γ

    IKK SIGNALING PANEL

    MCF-7 1 5 10 15 20 30 Days

    Day 1 Day 5 Day 10 Day 15 Day 20 Day 25 Day 30

    5uM T

    Day 1 Day 5 Day 3

    C

    APOPTOSIS PANEL

    MCF-7 1 5 10 15 20 30 Days

    PARP

    Cl-PARP

    Caspase9

    Cl-Caspase9

    pγ-H2AX (S139)

    y-H2AX

    GAPDH

    Bcl-2

    0

    5

    10

    15

    20

    0 2 4 6 8

    Cel

    l N

    um

    ber

    X

    Thousa

    nds

    Days

    MCF-7MCF-7 TamR TMCF-7 T

    *

    Fig. 3

    A

    RRM2

    RRM1

    RRM2B

    RIBONUCLEOTIDE REDUCTASE PANEL

    B

    C

    D

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • T-47D

    T+D

    T-47D

    TamR

    T+D

    Day 1 Day 15 Day 30 Day 1 Day 5 Day 10 Day 15 Day 20 Day 25 Day 30

    MCF-7

    TamR

    T+D

    MCF-7

    T+D

    pIKK-α/β (S176/180)

    pAKT (S473)

    pIkBα (S32)

    p-p65 (S536)

    pγ-H2AX (S139)

    RRM2

    RRM1

    RRM2B

    RIBONUCLEOTIDE REDUCTASE

    ER-α66

    ER-α36

    pER (S167)

    ER-β

    ESTROGEN RECEPTOR

    p53

    EGFR

    HER-2

    pERK1/2

    Total ERK1/2

    Total AKT

    GROWTH RECEPTOR

    p52

    p50

    p65

    RelB

    GAPDH

    C-Rel

    NF-κB SIGNALING

    IKK-δ

    IKK-β

    IKK-α

    Total IkBα

    IKK-ε

    IKK-γ

    IKK SIGNALING

    Total γ-H2AX

    PARP

    Cl-PARP

    Caspase9

    Cl-Caspase9

    GAPDH

    Bcl-2

    APOPTOSIS

    IKK-δ

    IKK-β

    IKK-α

    Total IkBα

    IKK-ε

    IKK-γ

    IKK SIGNALING

    p52

    p50

    p65

    RelB

    p-p65 (S536)

    GAPDH

    C-Rel

    NF-κB SIGNALING

    pIKK-α/β (S176/180)

    pAKT (S473)

    pIkBα (S32) Total γ-H2AX

    PARP

    Cl-PARP

    Caspase9

    Cl-Caspase9

    GAPDH

    Bcl-2

    APOPTOSIS

    pγ-H2AX (S139)

    RRM2

    RRM1

    RRM2B

    RIBONUCLEOTIDE REDUCTASE

    ER-α66

    ER-α36

    ER-β

    ESTROGEN RECEPTOR

    p53

    EGFR

    HER-2

    pERK1/2

    Total ERK1/2

    Total AKT

    GROWTH RECEPTOR

    pER (S167)

    -6

    -4

    -2

    0

    2

    4

    6

    MCF-7 MCF-7/R2.1 MCF-7 TamR T-47D T-47D TamR

    NF-κB reporter activity

    (Fold Change)

    C T D T+D

    *

    *

    *

    Fig. 4 A B

    E

    C D

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • A

    C

    B

    RRM2

    ER-a66

    ER-a36

    Ki67

    H&E

    T T+D425

    D

    Fig. 5

    ● ● ● ● **

    ** ** **

    * * *

    0

    20

    40

    60

    80

    100

    120

    140

    160

    7 14 21 28

    Me

    an T

    um

    or

    Vo

    lum

    e (

    mm

    3)

    Days

    CDTT+D425T+D200

    C D T T+D425

    RRM2

    ER-a66

    ER-a36

    GAPDH

    *

    * * **

    0.4

    0.6

    0.8

    1

    1.2

    1.4

    C D T T+D425

    Me

    an f

    old

    Co

    ntr

    ol

    RRM2 ER- 66 ER- 36a a

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • p=0.00065

    p=0.15 p=0.045

    -1

    1

    3

    5

    7

    9

    Grade1

    Grade2

    Grade3

    Rel

    ativ

    e m

    RN

    A E

    xpre

    ssio

    n

    (28)

    p=0.0009

    p=0.35 p=0.006

    -1

    1

    3

    5

    7

    Grade1

    Grade2

    Grade3

    Rel

    ativ

    e m

    RN

    A E

    xpre

    ssio

    n

    (26)

    A B C

    20

    70

    120%

    Op

    en a

    rea

    Cell Motility RRM2 - Didox RRM2 + Didox

    0

    1

    2

    O.D

    .

    Cell Invasion

    0

    1

    2

    O.D

    .

    Cell Migration

    E

    -40

    -30

    -20

    -10

    0

    10

    20

    30

    VC

    AM

    1

    MM

    P3

    SE

    LE

    MM

    P2

    PE

    CA

    M1

    FN

    1

    NC

    AM

    1

    MM

    P9

    ITG

    B3

    CN

    TN

    1

    CT

    GF

    LA

    MA

    3

    CD

    H1

    ICA

    M1

    CO

    L15A

    1

    CT

    NN

    A1

    Fo

    ld C

    han

    ge

    RRM2 -Didox RRM2 +Didox

    F

    G RRM2 ER-a36

    D

    Gra

    de

    1

    Gra

    de

    2

    Gra

    de

    3

    500 mm

    0

    50

    100

    150

    200

    250

    300

    Normal Grade 1

    Grade 2

    Grade 3

    Ave

    rage

    RR

    M2

    sta

    inin

    g (H

    -sco

    re)

    H

    0

    50

    100

    150

    200

    250

    300

    Normal Grade1

    Grade2

    Grade3

    Ave

    rage

    ER

    -a3

    6 (

    H-s

    core

    )

    I

    Fig. 6

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/

  • Published OnlineFirst September 2, 2015.Mol Cancer Ther Khyati N. Shah, Elizabeth A Wilson, Ritu Malla, et al. with Didox to circumvent tamoxifen resistance in breast cancer

    B activationκTargeting ribonucleotide reductase M2 and NF-

    Updated version

    10.1158/1535-7163.MCT-14-0689doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://mct.aacrjournals.org/content/suppl/2015/09/02/1535-7163.MCT-14-0689.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

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

    Subscriptions

    Reprints and

    [email protected] at

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

    Permissions

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

    .http://mct.aacrjournals.org/content/early/2015/09/02/1535-7163.MCT-14-0689To request permission to re-use all or part of this article, use this link

    on June 4, 2021. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 2, 2015; DOI: 10.1158/1535-7163.MCT-14-0689

    http://mct.aacrjournals.org/lookup/doi/10.1158/1535-7163.MCT-14-0689http://mct.aacrjournals.org/content/suppl/2015/09/02/1535-7163.MCT-14-0689.DC1http://mct.aacrjournals.org/cgi/alertsmailto:[email protected]://mct.aacrjournals.org/content/early/2015/09/02/1535-7163.MCT-14-0689http://mct.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure 4Figure 5Figure 6