Skip to main content
Erschienen in: BMC Cancer 1/2014

Open Access 01.12.2014 | Research article

Phospho-aspirin (MDC-22) inhibits breast cancer in preclinical animal models: an effect mediated by EGFR inhibition, p53 acetylation and oxidative stress

verfasst von: Liqun Huang, Chi C Wong, Gerardo G Mackenzie, Yu Sun, Ka Wing Cheng, Kvetoslava Vrankova, Ninche Alston, Nengtai Ouyang, Basil Rigas

Erschienen in: BMC Cancer | Ausgabe 1/2014

Abstract

Background

The anticancer properties of aspirin are restricted by its gastrointestinal toxicity and its limited efficacy. Therefore, we synthesized phospho-aspirin (PA-2; MDC-22), a novel derivative of aspirin, and evaluated its chemotherapeutic and chemopreventive efficacy in preclinical models of triple negative breast cancer (TNBC).

Methods

Efficacy of PA-2 was evaluated in human breast cancer cells in vitro, and in orthotopic and subcutaneous TNBC xenografts in nude mice. Mechanistic studies were also carried out to elucidate the mechanism of action of PA-2.

Results

PA-2 inhibited the growth of TNBC cells in vitro more potently than aspirin. Treatment of established subcutaneous TNBC xenografts (MDA-MB-231 and BT-20) with PA-2 induced a strong growth inhibitory effect, resulting in tumor stasis (79% and 90% inhibition, respectively). PA-2, but not aspirin, significantly prevented the development of orthotopic MDA-MB-231 xenografts (62% inhibition). Mechanistically, PA-2: 1) inhibited the activation of epidermal growth factor receptor (EGFR) and suppressed its downstream signaling cascades, including PI3K/AKT/mTOR and STAT3; 2) induced acetylation of p53 at multiple lysine residues and enhanced its DNA binding activity, leading to cell cycle arrest; and 3) induced oxidative stress by suppressing the thioredoxin system, consequently inhibiting the activation of the redox sensitive transcription factor NF-κB. These molecular alterations were observed in vitro and in vivo, demonstrating their relevance to the anticancer effect of PA-2.

Conclusions

Our findings demonstrate that PA-2 possesses potent chemotherapeutic efficacy against TNBC, and is also effective in its chemoprevention, warranting further evaluation as an anticancer agent.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-14-141) contains supplementary material, which is available to authorized users.

Competing interests

The authors have nothing to disclose except for BR, who has an equity position in Medicon Pharmaceuticals, Inc. and NO who is an employee for the same.

Authors’ contributions

LH conceived the study, participated in its design, carried out most of the in vitro and in vivo studies, analyzed the data and participated in the preparation of the manuscript. YS participated in the study design, performed the orthotopic transplantation of breast cancer cells and involved in the drafting the manucript; CCW, GGM, KWC participated in the study design, data analyses and writing of the manuscript. KV synthesized batches of PA-2, participated in the study design and data analysis. NA performed the luciferase imaging of breast tumors, participated in data collection and data analysis. NO performed the immunohistochemical analyses, participated in data collection and data analysis. BR participated in the study design, supervised the work, analyzed data and participated in writing the manuscript. All authors read and approved the final manuscript.
Abkürzungen
ADAM
A disintegrin and metalloproteinase
Akt
Protein kinase B
EGFR
Epidermal growth factor receptor
ER
Estrogen receptor
mTOR
Mammalian target of rapamycin
NSAID
Nonsteroidal anti-inflammatory drug
NF-ΚB
Nuclear factor kappa-light-chain-enhancer of activated B cells
PA-2
Phospho-aspirin-2
PI3K
Phosphoinositide 3-kinase
PR
Progesterone receptor
RONS
Reactive oxygen and nitrogen species
SIRT1
Sirtuin
STAT3
Signal transducer and activator of transcription 3
Trx-1
Thioredoxin-1
TrxR
Thioredoxin reductase
SC
Subcutaneous.

Background

Breast cancer is the second most common cause of female cancer-related deaths, with more than one million new cases diagnosed per year throughout the world [1]. Despite advances in its early detection, breast cancer remains a significant health problem. In particular, triple negative breast cancer (TNBC) is known to be more aggressive with poor prognosis, and is frequently associated with resistance to chemotherapeutic agents. Thus, novel agents capable of inhibiting TNBC are urgently needed.
Aspirin, a prototypical non-steroidal anti-inflammatory drug (NSAID), is the most widely used anti-inflammatory medication in the world [2, 3]. NSAIDs have a significant antineoplastic effect, which should be viewed, at least in part, in the context of the increasingly appreciated role of inflammation in cancer. Aspirin has been formally documented to be a chemopreventive agent against colon cancer [4, 5]. Epidemiological studies also support a role of aspirin in reducing the risk of breast cancer [6]. However, gastrointestinal toxicity caused by chronic aspirin use is a significant health concern. In order to reduce the toxicity and enhance the efficacy of aspirin, we synthesized phospho-aspirin (PA-2; MDC-22; Figure 1A), which consists of aspirin chemically modified at its –COOH group, the moiety accounting for its gastrointestinal toxicity [7, 8]. Indeed, as we have recently reported, the gastrointestinal toxicity of PA-2 in rats is much reduced compared to that of aspirin [9].
The epidermal growth factor receptor (EGFR) and p53 are key molecular determinants of TNBC [1012]. Aberrant activation of EGFR plays an important role in breast carcinogenesis via the sustained initiation of downstream cascades that promote cell survival and proliferation. Thus, EGFR is an attractive target for the development of cancer therapeutics [13]. On the other hand, the inactivation of p53, a potent tumor suppressor, is also a major contributor to breast cancer development [12]. Apart from its ability to block cell cycle progression and promote apoptosis, it is now appreciated that p53 also suppresses tumor development by modulating autophagy, cellular metabolism, angiogenesis, and metastasis [14]. This portends that the restoration of p53 function in tumors will be extremely beneficial, since it will not merely inhibit the growth of tumor cells but also obliterate the microenvironment required for tumor survival.
Herein, we examined the antineoplastic properties of PA-2 in TNBC in vitro and in vivo. PA-2 was much more potent than aspirin in inhibiting the growth of TNBC cells and strongly suppressed TNBC growth in subcutaneous and orthotopic xenograft models. Mechanistically, the antineoplastic effect of PA-2 is mediated through inhibition of EGFR, acetylation of p53 and induction of oxidative stress.

Methods

Reagents

PA-2 was provided by Medicon Pharmaceuticals, Inc., Setauket, NY. Aspirin were purchased from Sigma (St Louis, MO). For cell culture study, we prepared 500 mM stock solutions of both in DMSO. In all cell culture media, the final DMSO concentration was adjusted to 1%. All general solvents and reagents were of HPLC grade or of the highest grade commercially available. Antibodies against β-actin were from Sigma. All other antibodies were from Cell Signaling (Beverly, MA).

Cell culture

We used three human breast cancer cell lines: MDA-MB 231 (ER-, PR-, HER2/Neu-, EGFR+, and p53 mutant R280K), BT-20 ( ER-, PR-, HER2/Neu-, EGFR++, and p53 mutant K132G), and MDA-MB-468 (ER-, PR-, HER2/Neu-, EGFR++, and p53 mutant R2073H). All were obtained from the American Type Culture Collection (ATCC, Manassas, VA, and grown as monolayers in the specific medium and conditions suggested by ATCC. All cell lines were grown in our laboratory less than 6 months after their receipt and the cells studied were between passages 2-10.

Cell viability assay

We used an assay based on reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide dye (MTT), which was determined according to the manufacturer’s protocol (Promega, Madison, WI).

Cytokinetic analysis

For apoptosis, cells were seeded and treated with PA-2 for 24 h, trypsinized and stained with Annexin V-FITC (100X dilution; Invitrogen, Carlsbad, CA) and PI (0.5 μg/ml; Sigma, St Louis, MO), then analyzed by FACScaliber (BD Biosciences, San Jose, CA). To determine cell proliferation, we measured the incorporation of 5-bromo-2′-deoxyuridine (BrdU) into newly synthesized cellular DNA followed by the manufacture’s protocol (BD Biosciences), and cells were subjected to flow cytometric analysis. Cell cycle phase distribution was analyzed by flow cytometry as described [15].

Plasmid and siRNA transfection

EGFR, and SIRT1 plasmids were purchased from Addgene (Cambridge, MA). Transient transfection was performed with Lipofectamine 2000 (Invitrogen, Carlsbad, CA) following the manufacturer’s instructions.

Determination of reactive oxygen and nitrogen species (RONS)

After the indicated treatment, cells were collected by trypsinization, resuspended in 10 μM of 5-(and-6)-carboxy-2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA Invitrogen), or MitoSox Red (Invitrogen) or dihydroethidium (DHE, Sigma), incubated at 37°C for 30 min in the dark and their fluorescence intensity was determined by flow cytometry.

Urinary F2isoprostane assay

Urine was collected at the endpoint of treatment. Levels of F2 isoprostane and creatinine in urine were determined by ELISA (Oxford Biomedical Research, MA). F2-isoprostane values were normalized to creatinine levels.

Determination of TrxR reductase activity

After treatment, cells were lysed and TrxR activity was determined in the protein lysate using a commercially available kit, following the instructions of the manufacturer (Cayman Chemical, Ann Arbor, MI). In this assay, TrxR uses NADPH to reduce 5,5′-dithiobis-(2-nitrobenzoic acid) to 5-thio-2-nitrobenzoic acid (TNB). Glutathione (oxidized and reduced) was determined by the glutathione (GSH) reductase-coupled 5,5′-dithiobis(2-nitrobenzoic acid) assay [16].

Immunoblotting

After treatment with PA-2 as indicated, cells were scraped on ice, washed with ice-cold PBS and lysed in RIPA lysis buffer (Sigma). Protein concentration was determined using the Bradford method (Bio-Rad, Hercules, CA). Electrophoresis of cell lysates were performed on 10% SDS-polyacrylamide gel electrophoresis gels and protein was transferred onto nitrocellulose membranes as described [17];

Electrophoretic Mobility Shift Assay (EMSA)

Following treatment, nuclear fractions were isolated from 2 × 106 cells as described [16]. NF-κB, or p53 EMSA was performed according to The Thermo Scientific LightShift Chemiluminescent EMSA Kit (Rockford, IL) following the instructions of the manufacturer.

Efficacy studies in nude mouse breast xenografts and orthotopic model

All animal experiments were approved by the Institutional Animal Care and Use Committee.

Treatment protocol

Female Balb/C nude mice (Charles River Laboratories, Wilmington, MA) were inoculated subcutaneously into each of their flanks with 2.5-3 × 106 TNBC cells (MDA-MB-231 or BT-20) in Matrigel (BD Biosciences, Franklin Lakes, NJ). When the tumor reached approximately 100-150 mm3, animals were randomized into the control and treatment groups (n = 10/group). For MDA-MB-231 xenografts, the animals were treated with vehicle or PA-2 120 mg/kg p.o. in corn oil 5 times/wk. For BT-20 xenografts, animals were treated with vehicle or PA-2 300 mg/kg i.p. in corn oil 5 times/wk.

Prevention protocol

Female Balb/C nude mice were treated with PA-2 120 mg/kg or ASA 40 mg/kg p.o. in corn oil (equimolar) for 1 wk. Then, the mice were inoculated into the mammary fat pad with 1.0 × 106 MDA-MB-231 cells in Matrigel. Drug treatment was continued until the end of the study. Tumor volume was calculated as [length × width × (length + width/2) × 0.56]. At the end of treatment, animals were sacrificed and tumors were removed and weighed. To calculate tumor growth inhibition, we subtracted the baseline tumor volume from the final one.

Immunohistochemical analysis

Immunohistochemical staining for Ki-67, Dmp1 and phospho-NF-κB (p-p65, activated form of NF-κB) was performed on human breast xenograft tissue samples as previously described [18]. Apoptosis was determined by the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end-labeling (TUNEL) assay [19].

Statistical analysis

Results are expressed as mean ± SEM. Differences between groups were determined by one-factor analysis of variance followed by Tukey’s test for multiple comparisons. p < 0.05 was statistically significant.

Results

PA-2 inhibits the growth of human TNBC through a strong cytokinetic effect

We first compared the growth inhibitory effect of PA-2 and aspirin in a panel of TNBC cell lines. PA-2 inhibited cell growth more potently than aspirin in all the cell lines evaluated. The potency enhancement ranged between 5 and 7-fold in MDA-MB-231, MDA-MB-468, and BT-20 (Figure1A). PA-2 inhibited TNBC cell growth via a triple cytokinetic effect. In MDA-MB-231 and BT-20 cells, PA-2 a) inhibited cell proliferation by > 40% at 0.5 × IC50 and by > 80% at 1 × IC50; b) induced apoptosis by 1.6- to 12-fold over control at 1.5-and 2 × IC50; and c) suppressed the G1 to S cell cycle phase transition, leading to accumulation of cells in G1 phase by 14% at 0.5 × IC50 (Figure1B-D).
To assess the efficacy of PA-2 in vivo, we employed both subcutaneous and orthotopic TNBC xenografts in nude mice. Initially, we evaluated the chemotherapeutic effect of PA-2 on subcutaneous MDA-MB-231 and BT-20 xenografts. As shown in Figure 2A, PA-2 significantly inhibited MDA-MB-231 xenograft growth starting on day 8 of treatment until the end of the study (p < 0.001). At sacrifice, the tumor volume of vehicle was 309 ± 36 mm3 and that of PA-2 was 143 ± 16 mm3, representing a 79% tumor growth inhibition (p < 0.01). PA-2 also suppressed the growth of BT-20 xenografts (Figure 2B). After 28 days of treatment, the tumor volume of vehicle and PA-2 groups were 248 ± 27 mm3 and 157 ± 15 mm3, respectively (90% inhibition, p < 0.01).
We next evaluated the chemopreventive effect of PA-2 and compared it to aspirin, its parent compound. Following a prevention protocol, we treated nude mice bearing orthotopically implanted MDA-MB-231 xenografts with equimolar doses of PA-2 or aspirin starting 1 week before tumor implantation. On day 66 post-implantation, PA-2 inhibited the development of primary tumor in the mammary fat pads by 62% (p < 0.05; Figure 2C). In contrast, aspirin had no significant effect on breast tumor growth in this orthotopic model, consistent with previous findings [20].
We also determined cell proliferation and apoptosis in MDA-MB-231 xenografts in the treatment study (Figure 2A) using Ki-67 staining and TUNEL assay, respectively (Figure 2D). Compared to the vehicle, PA-2 inhibited cell proliferation by 44% (p < 0.01) and increased apoptosis by 3-fold (p < 0.002). This indicates that PA-2 also exerted a cytokinetic effect on TNBC xenografts in vivo.

PA-2 modulates the phosphorylation status of EGFR, p53 and NF-κB

To elucidate the mechanisms of action of PA-2, we performed antibody microarray analyses (Kinexus, Vancouver, Canada) on MDA-MB-231 cells treated with vehicle or PA-2 1.5 × IC50 for 2 h. This assay revealed pronounced changes in EGFR, p53 and NF-κB pathways following PA-2 treatment (Additional file 1: Table S1). Therefore, we further investigate the contribution of each of these pathways to the anti-cancer effect of PA-2.

PA-2 inhibits EGFR activation and its downstream signaling

EGFR is known to correlate with the progression of TNBC [11]. In MDA-MB-231 and BT-20 cells, PA-2 inhibited EGFR phosphorylation in a time-dependent manner, being evident as early as 1 h after treatment (Figure 3A). This observation was confirmed in vivo, where PA-2 reduced EGFR phosphorylation by 68% and 83% in MDA-MB-231 and BT-20 xenografts, respectively, compared to controls (p < 0.05, Figure 3A).
To determine the role of EGFR inhibition in the anticancer effect of PA-2, we transiently transfected MDA-MB-231 cells with an EGFR-overexpressing plasmid, and evaluated whether PA-2-induced cell death was affected. EGFR overexpression and activation of its downstream target p-PI3K was confirmed by western blot (Figure 3B). EGFR overexpression suppressed the induction of apoptosis by PA-2. Compared to mock transfected control, EGFR-overexpressing MDA-MB-231 cells have 2.5-fold reduction in the annexin V (+) fraction after treatment with PA-2 2xIC50 (Figure 3B). This indicates that EGFR inhibition is an important mechanism of action of PA-2, and the reversal of this effect mediates drug resistance.
An important upstream regulator of EGFR phosphorylation is ADAM17, which activates EGFR through a ligand cleavage mechanism [21]. We assessed the effect of PA-2 on ADAM proteins. As shown in Figure 2C, in MDA-MB231 and BT-20 cells, PA-2 reduced the expression of ADAM17. Moreover, PA-2 suppressed the levels of ADAM17 in MDA-MB-231 orthotopic xenografts.
Inhibition of EGFR activation resulted in a potent inhibitory effect on its downstream signaling cascades, STAT3 and PI3K/Akt pathways. PA-2 reduced STAT3 phosphorylation in MDA-MB-231 and BT-20 cells (Figure 4A). PA-2 also suppressed the levels of p-PI3K and p-Akt in these cells in vitro (Figure 4B) and in MDA-MB-231 xenografts (Figure 4C). In addition, the downstream targets of PI3K/Akt pathway, including p-mTOR, p-4E-BP1 and p-70S6K1, were reduced after prolonged (16h) PA-2 treatment (Figure 4B). Hence, PA-2 triggered a temporal suppression of EGFR signaling cascades in TNBC.

PA-2 induces acetylation of p53 and cell cycle arrest

The tumor suppressor gene p53 is frequently inactivated in TNBC [22]. In MDA-MB-231 and BT-20 cells, PA-2 enhanced the DNA-binding activity of p53 in a concentration-dependent manner (Figure 5A). PA-2 did not appear to alter the nuclear shuttling of p53 (Figure 5A). On the other hand, immunoprecipitation showed that PA-2 significantly reduced the binding of p53 to murine double minute 2 (MDM2) (Figure 5B). Dissociation of p53 from MDM2, which otherwise binds to p53 and represses its transcriptional activity [23], may therefore contribute to the induction of p53 DNA binding activity by PA-2 in TNBC cells. The activation of p53 by PA-2 was consequential, as PA-2 blocked G1 to S cell cycle transition (Figure 1D) and up-regulated p21 in TNBC cells in vitro and in MDA-MB-231 xenografts (Figure 5C).
Acetylation of p53 at lysine residues is critical for its stability and transcriptional activity [24]. Given that the aspirin moiety of PA-2 contains an acetyl group capable of acetylating multiple proteins in cancer cells [25], we examined the effect of PA-2 on the acetylation status of p53. In MDA-MB-231 cells, PA-2 induced p53 acetylation at three distinct lysine residues (K373, K379 and K382) in a time-dependent manner; while in BT-20 cells PA-2 induced acetylation at K373 and K379 residues (Figure 6A). In MDA-MB-231 and BT-20 xenografts, treatment with PA-2 increased p53 acetylation at K382 and K373 residues, respectively (Figure 6A).
To further assess the role of p53 acetylation in cell death induction by PA-2, we overexpressed in MDA-MB-231 cells SIRT1, which negatively regulates p53 through its de-acetylation [26, 27]. Overexpression of SIRT1 blocked the ability of PA-2 to acetylate p53 at the K382 residue (Figure 6B). Importantly, SIRT1 overexpression attenuated the induction of apoptosis in response to PA-2 by 71%, indicating that PA-2 induces apoptosis, at least in part, by a p53 acetylation-dependent mechanism (Figure 6B).
PA-2 may also regulate p53 independently of acetylation. PA-2 significantly enhanced the expression of Dmp1, a tumor suppressor that induces p53-dependent cell cycle arrest by directly binding to its promoter [28]. Such an effect is observed in TNBC cells in vitro. In MDA-MB-231 xenografts, PA-2 treatment increased Dmp1 expression by 57% (p < 0.02) compared to the control group (Figure 6C).

PA-2 induces RONS levels, inhibits the thioredoxin system and NF-κB activation

RONS play a significant role in the action of phospho-NSAIDs [29]. We determined the effect of PA-2 using various molecular probes: DCFDA (general RONS), DHE (cytoplasmic O2), and MitoSOX Red (mitochondrial O2). Compared to control, PA-2 1.5 × IC50 increased DCFDA by 76%, DHE by 51% and MitoSOX Red by 51% in MDA-MB-231 cells (Figure 7A). N-acetylcysteine (10 mM), a ROS scavenger, partly blocked ROS induction by 26% in MDA-MB-231 cells (Figure 7A). In BT-20 cells, PA-2 increased MitoSOX Red by 25%. PA-2 1-1.5 × IC50 also decreased the level of glutathione, a major cellular antioxidant. Co-incubation of PA-2 with BSO, an inhibitor of GSH synthesis, synergistically induced RONS levels and inhibited cell growth (Figure 7B).
To assess the effect of PA-2 on RONS in vivo, we measured urinary 15-F2t-isoprostane, a marker of oxidative stress [30, 31], in the mice bearing TNBC xenografts. In orthotopic MDA-MB-231 xenografts (Figure 2C), 15-F2t-isoprostane levels on day 25 were 28.7 ± 3.3 ng/mg creatinine in controls and 40.9 ± 2.2 ng/mg creatinine in the PA-2 group, representing a nearly 40% increase (p < 0.05) (Figure 7C). In contrast, aspirin had no significant effect (p = 0.6). In BT-20 xenografts, PA-2 treatment increased 15-F2t-isoprostane levels by over 3-fold over the control (p < 0.006). Hence, PA-2, but not aspirin, induced RONS in vivo.
The thioredoxin (Trx) system, composed of thioredoxin reductase (TrxR) and Trx-1, plays an important role in redox homeostasis by reducing oxidized proteins; the latter is overexpressed in TNBC [32]. PA-2 inhibited TrxR activity in MDA-MB-231 cells in cell culture and MDA-MB-231 xenografts by 54% and 41%, respectively (p < 0.02-0.04), without affecting its levels (Figure 8A). PA-2 also significantly reduced Trx-1 levels in MDA-MB-231 cells after 1h treatment with 1.5 × IC50 PA-2 (Figure 8A). These results indicate that PA-2 targets major components of the Trx system.
The Trx system is closely linked with the NF-κB signaling pathway. Trx-1 enhances DNA binding of NF-κB by reducing the intermolecular Cys62 -S-S- bond of its p50 subunit [33]. We thus examined the effect on PA-2 on NF-κB activation. Consistent with its inhibitory effect on Trx-1 expression, PA-2 also inhibited NF-κB-DNA binding in a concentration-dependent manner in MDA-MB-231 and BT-20 cells (Figure 8B). A similar inhibition was observed in MDA-MB-231 xenografts. PA-2 reduced the levels of activated p-p65 by 44% (p < 0.009), compared to the control group (Figure 8B).

Discussion

Our data demonstrate that PA-2, a novel derivative of aspirin, effectively inhibits TNBC in preclinical models and is much more potent than aspirin, its parent compound. The anticancer activity of PA-2 is associated with a pronounced effect on a) EGFR activation; b) p53 acetylation and c) RONS induction. These signaling effects of PA-2 culminate in the substantial inhibition of cell proliferation and induction of apoptosis, the net effect of which is a strong reduction in TNBC xenograft growth in vivo.
Efficacy and safety are the prime considerations in the evaluation of anticancer agents [34], and the latter is especially paramount in chemoprevention. Extensive epidemiological and clinical evidence supports a beneficial role of aspirin in the prevention of breast, colon, and lung cancers [35]; however, its gastrointestinal toxicity is limiting. Our results demonstrated that PA-2, with the novel phospho-modification, is superior to aspirin both in terms of efficacy in TNBC, as well as safety [9]. PA-2, but not aspirin, administered in a chemoprevention protocol slowed the development of orthotopic MDA-MB-231 xenografts.
PA-2 is highly efficacious when used in the treatment setting and its chemotherapeutic effect is even stronger than its chemopreventive effect. PA-2 potently inhibits TNBC in vivo, almost completely arresting the growth of both MDA-MB-231 and BT-20 xenografts. Consistent with its strong chemotherapeutic efficacy, PA-2 induced a profound cytokinetic effect involving inhibition of cell proliferation and induction of apoptosis. Hence, PA-2 is a promising anticancer candidate that merits further evaluation.
Our work identified EGFR, p53 and RONS as the major signaling mechanisms (Figure 9) involved in eliciting the growth inhibitory effect of PA-2. EGFR is overexpressed in 16–48% of breast cancers and its expression is associated with poor prognosis [36, 37]. In particular, EGFR is frequently overexpressed in TNBC, a subset of breast cancer that is characterized by their unique molecular profile, aggressive behavior and distinct patterns of metastasis [38]. PA-2 is an inhibitor of EGFR phosphorylation in vitro and in vivo. The inhibitory effect of PA-2 appears to be mediated by a novel mechanism involving the inhibition of ADAM17, a major protease that controls availability of EGFR ligands [39]. A key consequence of PA-2-induced inhibition of EGFR is the suppression of pro-survival STAT3 and PI3K and the sequential inactivation of their downstream signaling propagation, thereby causing growth inhibition and cell death.
Mutations of p53 are exceptionally frequent in TNBC (>80%) and its inactivation predicts poor survival in TNBC patients [10, 12]. Activation-inactivation of p53 depends on a repertoire of post-translational modifications [40], including phosphorylation and acetylation. p53 acetylation was found to be indispensable for its activation, as it destabilizes the p53-MDM2 interaction, thereby abrogating MDM2-mediated transcription repression [24]. Here, we show that PA-2 acetylates p53 in vitro and in vivo and disrupts its association with MDM2, which in turn, enhances p53-DNA binding activity. Forced deacetylation of p53 partly attenuated cell death induction by PA-2, indicating an important role of p53 acetylation in mediating the effect of PA-2. PA-2 additionally regulates p53 independently of acetylation. Dmp1 is a transcription factor that physically interacts with p53 [41]. Dmp1-p53 binding antagonizes the ubiquitination of p53 byMDM-2 and promotes the nuclear translocation of p53 [28]. PA-2 enhanced the expression of Dmp1 in TNBC cells and in xenografts, which may further contribute to the robust p53 activation triggered by PA-2.
The induction of oxidative stress plays a key role in the anticancer effect of structurally related phospho-NSAIDs [9, 42]. PA-2 induces oxidative stress in TNBC cells in culture and in xenografts by compromising cellular antioxidant defense mechanisms. First, PA-2 suppressed the Trx system, with both TrxR and Trx-1 affected in a significant way. TrxR and Trx-1 are critical components of the cellular redox system [43]. Trx-1 reduces client proteins oxidized by RONS, with itself undergoing oxidation, while TrxR regenerates the reduced Trx-1. In TNBC cells and xenografts, PA-2 inhibited TrxR activity and decreased the expression of Trx-1. Second, PA-2 significantly reduced intracellular levels of GSH, the major chemical antioxidant of mammalian cells. Induction of oxidative stress has major repercussions on redox- and Trx-dependent signaling cascades, as exemplified by the inhibitory effect on NF-кB. NF-кB is constitutively active in TNBC [44], and its aberrant activation is linked to inflammation and cancer. Induction of oxidative stress and the subsequent inhibition of NF-кB by PA-2 may mediate part of its growth inhibitory effect.
It is of interest that PA-2 has superior efficacy compared to conventional aspirin, the starting compound for its synthesis. The reasons for this difference are not entirely clear, but as a new chemical entity, PA-2 is expected to have distinct properties from its parent compound. Perhaps the most relevant property of these two compounds is their ability to induce oxidative stress, an effect that accounts for much of the anticancer efficacy of phospho-NSAIDs, including phospho-aspirin [29]. Indeed, PA-2 caused robust oxidative stress in mice, reflected in enhanced urinary levels of the biomarker F2-isoprostane. In contrast, aspirin failed to produce such an effect, and this difference explains to a large extent their differential efficacy. Additional mechanistic differences might also contribute, especially with regards to effects on cell signaling cascades.

Conclusions

Our work indicates that PA-2 possesses chemotherapeutic and chemopreventive efficacy against TNBC in preclinical models, and establishes inhibition of EGFR, acetylation of p53 and induction of oxidative stress as critical mediators of its mechanism of action (Figure 9). The multi-targeted nature of PA-2 towards the dysregulated signaling cascades in TNBC further suggests the notion that PA-2 may be a promising therapeutic option, either alone or in combination with other therapies.

Acknowledgement

Grant support: NIH grants R01-CA139453, R01CA13945402 and 1N01CN43302WA22 and HHSN261201000109C.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​ ) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors have nothing to disclose except for BR, who has an equity position in Medicon Pharmaceuticals, Inc. and NO who is an employee for the same.

Authors’ contributions

LH conceived the study, participated in its design, carried out most of the in vitro and in vivo studies, analyzed the data and participated in the preparation of the manuscript. YS participated in the study design, performed the orthotopic transplantation of breast cancer cells and involved in the drafting the manucript; CCW, GGM, KWC participated in the study design, data analyses and writing of the manuscript. KV synthesized batches of PA-2, participated in the study design and data analysis. NA performed the luciferase imaging of breast tumors, participated in data collection and data analysis. NO performed the immunohistochemical analyses, participated in data collection and data analysis. BR participated in the study design, supervised the work, analyzed data and participated in writing the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Siegel R, Naishadham D, Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 2012, 62 (1): 10-29. 10.3322/caac.20138.CrossRefPubMed Siegel R, Naishadham D, Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 2012, 62 (1): 10-29. 10.3322/caac.20138.CrossRefPubMed
3.
Zurück zum Zitat Stanley P, Hegedus R: Aspirin–the first hundred years. Biologist (London). 2000, 47 (5): 269-271. Stanley P, Hegedus R: Aspirin–the first hundred years. Biologist (London). 2000, 47 (5): 269-271.
4.
Zurück zum Zitat Baron JA: What now for aspirin and cancer prevention?. J Natl Cancer Inst. 2004, 96 (1): 4-5. 10.1093/jnci/djh027.CrossRefPubMed Baron JA: What now for aspirin and cancer prevention?. J Natl Cancer Inst. 2004, 96 (1): 4-5. 10.1093/jnci/djh027.CrossRefPubMed
5.
Zurück zum Zitat Baron JA, Cole BF, Sandler RS, Haile RW, Ahnen D, Bresalier R, McKeown-Eyssen G, Summers RW, Rothstein R, Burke CA, Snover DC, Church TR, Allen JI, Beach M, Beck GJ, Bond JH, Byers T, Greenberg ER, Mandel JS, Marcon N, Mott LA, Pearson L, Saibil F, van Stolk RU: A randomized trial of aspirin to prevent colorectal adenomas. N Engl J Med. 2003, 348 (10): 891-899. 10.1056/NEJMoa021735.CrossRefPubMed Baron JA, Cole BF, Sandler RS, Haile RW, Ahnen D, Bresalier R, McKeown-Eyssen G, Summers RW, Rothstein R, Burke CA, Snover DC, Church TR, Allen JI, Beach M, Beck GJ, Bond JH, Byers T, Greenberg ER, Mandel JS, Marcon N, Mott LA, Pearson L, Saibil F, van Stolk RU: A randomized trial of aspirin to prevent colorectal adenomas. N Engl J Med. 2003, 348 (10): 891-899. 10.1056/NEJMoa021735.CrossRefPubMed
6.
Zurück zum Zitat Marshall SF, Bernstein L, Anton-Culver H, Deapen D, Horn-Ross PL, Mohrenweiser H, Peel D, Pinder R, Purdie DM, Reynolds P, Stram D, West D, Wright WE, Ziogas A, Ross RK: Nonsteroidal anti-inflammatory drug use and breast cancer risk by stage and hormone receptor status. J Natl Cancer Inst. 2005, 97 (11): 805-812. 10.1093/jnci/dji140.CrossRefPubMed Marshall SF, Bernstein L, Anton-Culver H, Deapen D, Horn-Ross PL, Mohrenweiser H, Peel D, Pinder R, Purdie DM, Reynolds P, Stram D, West D, Wright WE, Ziogas A, Ross RK: Nonsteroidal anti-inflammatory drug use and breast cancer risk by stage and hormone receptor status. J Natl Cancer Inst. 2005, 97 (11): 805-812. 10.1093/jnci/dji140.CrossRefPubMed
7.
Zurück zum Zitat Piazza GA, Keeton AB, Tinsley HN, Gary BD, Whitt JD, Mathew B, Thaiparambil J, Coward L, Gorman G, Li Y, Sani B, Hobrath JV, Maxuitenko YY, Reynolds RC: A novel sulindac derivative that does not inhibit cyclooxygenases but potently inhibits colon tumor cell growth and induces apoptosis with antitumor activity. Cancer Prev Res (Phila). 2009, 2 (6): 572-580. 10.1158/1940-6207.CAPR-09-0001.CrossRef Piazza GA, Keeton AB, Tinsley HN, Gary BD, Whitt JD, Mathew B, Thaiparambil J, Coward L, Gorman G, Li Y, Sani B, Hobrath JV, Maxuitenko YY, Reynolds RC: A novel sulindac derivative that does not inhibit cyclooxygenases but potently inhibits colon tumor cell growth and induces apoptosis with antitumor activity. Cancer Prev Res (Phila). 2009, 2 (6): 572-580. 10.1158/1940-6207.CAPR-09-0001.CrossRef
8.
Zurück zum Zitat Wong CC, Cheng KW, Xie G, Zhou D, Zhu CH, Constantinides PP, Rigas B: Carboxylesterases 1 and 2 hydrolyze phospho-nonsteroidal anti-inflammatory drugs: relevance to their pharmacological activity. J Pharmacol Exp Ther. 2012, 340 (2): 422-432. 10.1124/jpet.111.188508.CrossRefPubMedPubMedCentral Wong CC, Cheng KW, Xie G, Zhou D, Zhu CH, Constantinides PP, Rigas B: Carboxylesterases 1 and 2 hydrolyze phospho-nonsteroidal anti-inflammatory drugs: relevance to their pharmacological activity. J Pharmacol Exp Ther. 2012, 340 (2): 422-432. 10.1124/jpet.111.188508.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Huang L, Mackenzie G, Ouyang N, Sun Y, Xie G, Johnson F, Komninou D, Rigas B: The novel phospho-non-steroidal anti-inflammatory drugs, OXT-328, MDC-22 and MDC-917, inhibit adjuvant-induced arthritis in rats. Br J Pharmacol. 2011, 162 (7): 1521-1533. 10.1111/j.1476-5381.2010.01162.x.CrossRefPubMedPubMedCentral Huang L, Mackenzie G, Ouyang N, Sun Y, Xie G, Johnson F, Komninou D, Rigas B: The novel phospho-non-steroidal anti-inflammatory drugs, OXT-328, MDC-22 and MDC-917, inhibit adjuvant-induced arthritis in rats. Br J Pharmacol. 2011, 162 (7): 1521-1533. 10.1111/j.1476-5381.2010.01162.x.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Price JT, Tiganis T, Agarwal A, Djakiew D, Thompson EW: Epidermal growth factor promotes MDA-MB-231 breast cancer cell migration through a phosphatidylinositol 3′-kinase and phospholipase C-dependent mechanism. Cancer Res. 1999, 59 (21): 5475-5478.PubMed Price JT, Tiganis T, Agarwal A, Djakiew D, Thompson EW: Epidermal growth factor promotes MDA-MB-231 breast cancer cell migration through a phosphatidylinositol 3′-kinase and phospholipase C-dependent mechanism. Cancer Res. 1999, 59 (21): 5475-5478.PubMed
12.
Zurück zum Zitat Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature. 2012, 490 (7418): 61-70. 10.1038/nature11412.CrossRef Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature. 2012, 490 (7418): 61-70. 10.1038/nature11412.CrossRef
13.
Zurück zum Zitat Dancey JE, Freidlin B: Targeting epidermal growth factor receptor–are we missing the mark?. Lancet. 2003, 362 (9377): 62-64. 10.1016/S0140-6736(03)13810-X.CrossRefPubMed Dancey JE, Freidlin B: Targeting epidermal growth factor receptor–are we missing the mark?. Lancet. 2003, 362 (9377): 62-64. 10.1016/S0140-6736(03)13810-X.CrossRefPubMed
14.
Zurück zum Zitat Vousden KH, Prives C: Blinded by the light: the growing complexity of p53. Cell. 2009, 137 (3): 413-431. 10.1016/j.cell.2009.04.037.CrossRefPubMed Vousden KH, Prives C: Blinded by the light: the growing complexity of p53. Cell. 2009, 137 (3): 413-431. 10.1016/j.cell.2009.04.037.CrossRefPubMed
15.
Zurück zum Zitat Zhao W, Mackenzie GG, Murray OT, Zhang Z, Rigas B: Phosphoaspirin (MDC-43), a novel benzyl ester of aspirin, inhibits the growth of human cancer cell lines more potently than aspirin: a redox-dependent effect. Carcinogenesis. 2009, 30 (3): 512-519. 10.1093/carcin/bgp015.CrossRefPubMedPubMedCentral Zhao W, Mackenzie GG, Murray OT, Zhang Z, Rigas B: Phosphoaspirin (MDC-43), a novel benzyl ester of aspirin, inhibits the growth of human cancer cell lines more potently than aspirin: a redox-dependent effect. Carcinogenesis. 2009, 30 (3): 512-519. 10.1093/carcin/bgp015.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Mackenzie GG, Sun Y, Huang L, Xie G, Ouyang N, Gupta RC, Johnson F, Komninou D, Kopelovich L, Rigas B: Phospho-sulindac (OXT-328), a novel sulindac derivative, is safe and effective in colon cancer prevention in mice. Gastroenterology. 2010, 139 (4): 1320-1332. 10.1053/j.gastro.2010.06.044.CrossRefPubMedPubMedCentral Mackenzie GG, Sun Y, Huang L, Xie G, Ouyang N, Gupta RC, Johnson F, Komninou D, Kopelovich L, Rigas B: Phospho-sulindac (OXT-328), a novel sulindac derivative, is safe and effective in colon cancer prevention in mice. Gastroenterology. 2010, 139 (4): 1320-1332. 10.1053/j.gastro.2010.06.044.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Hundley TR, Gilfillan AM, Tkaczyk C, Andrade MV, Metcalfe DD, Beaven MA: Kit and FcepsilonRI mediate unique and convergent signals for release of inflammatory mediators from human mast cells. Blood. 2004, 104 (8): 2410-2417. 10.1182/blood-2004-02-0631.CrossRefPubMed Hundley TR, Gilfillan AM, Tkaczyk C, Andrade MV, Metcalfe DD, Beaven MA: Kit and FcepsilonRI mediate unique and convergent signals for release of inflammatory mediators from human mast cells. Blood. 2004, 104 (8): 2410-2417. 10.1182/blood-2004-02-0631.CrossRefPubMed
18.
Zurück zum Zitat Ouyang N, Williams JL, Rigas B: NO-donating aspirin isomers downregulate peroxisome proliferator-activated receptor (PPAR){delta} expression in APCmin/+ mice proportionally to their tumor inhibitory effect: Implications for the role of PPAR{delta} in carcinogenesis. Carcinogenesis. 2006, 27 (2): 232-239. 10.1093/carcin/bgi221.CrossRefPubMed Ouyang N, Williams JL, Rigas B: NO-donating aspirin isomers downregulate peroxisome proliferator-activated receptor (PPAR){delta} expression in APCmin/+ mice proportionally to their tumor inhibitory effect: Implications for the role of PPAR{delta} in carcinogenesis. Carcinogenesis. 2006, 27 (2): 232-239. 10.1093/carcin/bgi221.CrossRefPubMed
19.
Zurück zum Zitat Rigas B, Kozoni V: The novel phenylester anticancer compounds: Study of a derivative of aspirin (phoshoaspirin). Int J Oncol. 2008, 32 (1): 97-100.PubMed Rigas B, Kozoni V: The novel phenylester anticancer compounds: Study of a derivative of aspirin (phoshoaspirin). Int J Oncol. 2008, 32 (1): 97-100.PubMed
20.
Zurück zum Zitat Likhite V: Aspirin and Breast Cancer: Studies In Mice. Central Reginoal Meeting of the American Chemical Society. 2009, Cleveland, Ohio Likhite V: Aspirin and Breast Cancer: Studies In Mice. Central Reginoal Meeting of the American Chemical Society. 2009, Cleveland, Ohio
21.
Zurück zum Zitat Baumgart A, Seidl S, Vlachou P, Michel L, Mitova N, Schatz N, Specht K, Koch I, Schuster T, Grundler R, Kremer M, Fend F, Siveke JT, Peschel C, Duyster J, Dechow T: ADAM17 regulates epidermal growth factor receptor expression through the activation of Notch1 in non-small cell lung cancer. Cancer Res. 2010, 70 (13): 5368-5378. 10.1158/0008-5472.CAN-09-3763.CrossRefPubMed Baumgart A, Seidl S, Vlachou P, Michel L, Mitova N, Schatz N, Specht K, Koch I, Schuster T, Grundler R, Kremer M, Fend F, Siveke JT, Peschel C, Duyster J, Dechow T: ADAM17 regulates epidermal growth factor receptor expression through the activation of Notch1 in non-small cell lung cancer. Cancer Res. 2010, 70 (13): 5368-5378. 10.1158/0008-5472.CAN-09-3763.CrossRefPubMed
22.
Zurück zum Zitat Soussi T, Lozano G: p53 mutation heterogeneity in cancer. Biochem Biophys Res Commun. 2005, 331 (3): 834-842. 10.1016/j.bbrc.2005.03.190.CrossRefPubMed Soussi T, Lozano G: p53 mutation heterogeneity in cancer. Biochem Biophys Res Commun. 2005, 331 (3): 834-842. 10.1016/j.bbrc.2005.03.190.CrossRefPubMed
23.
Zurück zum Zitat Meek DW, Anderson CW: Posttranslational modification of p53: cooperative integrators of function. Cold Spring Harb Perspect Biol. 2009, 1 (6): a000950-CrossRefPubMedPubMedCentral Meek DW, Anderson CW: Posttranslational modification of p53: cooperative integrators of function. Cold Spring Harb Perspect Biol. 2009, 1 (6): a000950-CrossRefPubMedPubMedCentral
24.
25.
Zurück zum Zitat Marimuthu S, Chivukula RS, Alfonso LF, Moridani M, Hagen FK, Bhat GJ: Aspirin acetylates multiple cellular proteins in HCT-116 colon cancer cells: identification of novel targets. Int J Oncol. 2011, 39 (5): 1273-1283.PubMed Marimuthu S, Chivukula RS, Alfonso LF, Moridani M, Hagen FK, Bhat GJ: Aspirin acetylates multiple cellular proteins in HCT-116 colon cancer cells: identification of novel targets. Int J Oncol. 2011, 39 (5): 1273-1283.PubMed
26.
Zurück zum Zitat Ito A, Kawaguchi Y, Lai CH, Kovacs JJ, Higashimoto Y, Appella E, Yao TP: MDM2-HDAC1-mediated deacetylation of p53 is required for its degradation. EMBO J. 2002, 21 (22): 6236-6245. 10.1093/emboj/cdf616.CrossRefPubMedPubMedCentral Ito A, Kawaguchi Y, Lai CH, Kovacs JJ, Higashimoto Y, Appella E, Yao TP: MDM2-HDAC1-mediated deacetylation of p53 is required for its degradation. EMBO J. 2002, 21 (22): 6236-6245. 10.1093/emboj/cdf616.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Solomon JM, Pasupuleti R, Xu L, McDonagh T, Curtis R, DiStefano PS, Huber LJ: Inhibition of SIRT1 catalytic activity increases p53 acetylation but does not alter cell survival following DNA damage. Mol Cell Biol. 2006, 26 (1): 28-38. 10.1128/MCB.26.1.28-38.2006.CrossRefPubMedPubMedCentral Solomon JM, Pasupuleti R, Xu L, McDonagh T, Curtis R, DiStefano PS, Huber LJ: Inhibition of SIRT1 catalytic activity increases p53 acetylation but does not alter cell survival following DNA damage. Mol Cell Biol. 2006, 26 (1): 28-38. 10.1128/MCB.26.1.28-38.2006.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Taneja P, Maglic D, Kai F, Sugiyama T, Kendig RD, Frazier DP, Willingham MC, Inoue K: Critical roles of DMP1 in human epidermal growth factor receptor 2/neu-Arf-p53 signaling and breast cancer development. Cancer Res. 2010, 70 (22): 9084-9094. 10.1158/0008-5472.CAN-10-0159.CrossRefPubMedPubMedCentral Taneja P, Maglic D, Kai F, Sugiyama T, Kendig RD, Frazier DP, Willingham MC, Inoue K: Critical roles of DMP1 in human epidermal growth factor receptor 2/neu-Arf-p53 signaling and breast cancer development. Cancer Res. 2010, 70 (22): 9084-9094. 10.1158/0008-5472.CAN-10-0159.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Sun Y, Huang L, Mackenzie GG, Rigas B: Oxidative stress mediates through apoptosis the anticancer effect of phospho-nonsteroidal anti-inflammatory drugs: implications for the role of oxidative stress in the action of anticancer agents. J Pharmacol Exp Ther. 2011, 338 (3): 775-783. 10.1124/jpet.111.183533.CrossRefPubMedPubMedCentral Sun Y, Huang L, Mackenzie GG, Rigas B: Oxidative stress mediates through apoptosis the anticancer effect of phospho-nonsteroidal anti-inflammatory drugs: implications for the role of oxidative stress in the action of anticancer agents. J Pharmacol Exp Ther. 2011, 338 (3): 775-783. 10.1124/jpet.111.183533.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Basu S: F2-isoprostanes in human health and diseases: from molecular mechanisms to clinical implications. Antioxid Redox Signal. 2008, 10 (8): 1405-1434. 10.1089/ars.2007.1956.CrossRefPubMed Basu S: F2-isoprostanes in human health and diseases: from molecular mechanisms to clinical implications. Antioxid Redox Signal. 2008, 10 (8): 1405-1434. 10.1089/ars.2007.1956.CrossRefPubMed
31.
Zurück zum Zitat Tacconelli S, Capone ML, Patrignani P: Measurement of 8-iso-prostaglandin F2alpha in biological fluids as a measure of lipid peroxidation. Methods Mol Biol. 2010, 644: 165-178. 10.1007/978-1-59745-364-6_14.CrossRefPubMed Tacconelli S, Capone ML, Patrignani P: Measurement of 8-iso-prostaglandin F2alpha in biological fluids as a measure of lipid peroxidation. Methods Mol Biol. 2010, 644: 165-178. 10.1007/978-1-59745-364-6_14.CrossRefPubMed
32.
Zurück zum Zitat Mukherjee A, Martin SG: The thioredoxin system: a key target in tumour and endothelial cells. Br J Radiol. 2008, 81 (Spec No 1): S57-S68.CrossRefPubMed Mukherjee A, Martin SG: The thioredoxin system: a key target in tumour and endothelial cells. Br J Radiol. 2008, 81 (Spec No 1): S57-S68.CrossRefPubMed
33.
Zurück zum Zitat Matthews JR, Wakasugi N, Virelizier JL, Yodoi J, Hay RT: Thioredoxin regulates the DNA binding activity of NF-kappa B by reduction of a disulphide bond involving cysteine 62. Nucleic Acids Res. 1992, 20 (15): 3821-3830. 10.1093/nar/20.15.3821.CrossRefPubMedPubMedCentral Matthews JR, Wakasugi N, Virelizier JL, Yodoi J, Hay RT: Thioredoxin regulates the DNA binding activity of NF-kappa B by reduction of a disulphide bond involving cysteine 62. Nucleic Acids Res. 1992, 20 (15): 3821-3830. 10.1093/nar/20.15.3821.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Wong CC, Cheng KW, Rigas B: Preclinical predictors of anticancer drug efficacy: critical assessment with emphasis on whether nanomolar potency should be required of candidate agents. J Pharmacol Exp Ther. 2012, 341 (3): 572-578. 10.1124/jpet.112.191957.CrossRefPubMedPubMedCentral Wong CC, Cheng KW, Rigas B: Preclinical predictors of anticancer drug efficacy: critical assessment with emphasis on whether nanomolar potency should be required of candidate agents. J Pharmacol Exp Ther. 2012, 341 (3): 572-578. 10.1124/jpet.112.191957.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Cuzick J, Otto F, Baron JA, Brown PH, Burn J, Greenwald P, Jankowski J, La Vecchia C, Meyskens F, Senn HJ, Thun M: Aspirin and non-steroidal anti-inflammatory drugs for cancer prevention: an international consensus statement. Lancet Oncol. 2009, 10 (5): 501-507. 10.1016/S1470-2045(09)70035-X.CrossRefPubMed Cuzick J, Otto F, Baron JA, Brown PH, Burn J, Greenwald P, Jankowski J, La Vecchia C, Meyskens F, Senn HJ, Thun M: Aspirin and non-steroidal anti-inflammatory drugs for cancer prevention: an international consensus statement. Lancet Oncol. 2009, 10 (5): 501-507. 10.1016/S1470-2045(09)70035-X.CrossRefPubMed
36.
Zurück zum Zitat Tsutsui S, Kataoka A, Ohno S, Murakami S, Kinoshita J, Hachitanda Y: Prognostic and predictive value of epidermal growth factor receptor in recurrent breast cancer. Clin Cancer Res. 2002, 8 (11): 3454-3460.PubMed Tsutsui S, Kataoka A, Ohno S, Murakami S, Kinoshita J, Hachitanda Y: Prognostic and predictive value of epidermal growth factor receptor in recurrent breast cancer. Clin Cancer Res. 2002, 8 (11): 3454-3460.PubMed
37.
Zurück zum Zitat Tsutsui S, Ohno S, Murakami S, Hachitanda Y, Oda S: Prognostic value of epidermal growth factor receptor (EGFR) and its relationship to the estrogen receptor status in 1029 patients with breast cancer. Breast Cancer Res Treat. 2002, 71 (1): 67-75. 10.1023/A:1013397232011.CrossRefPubMed Tsutsui S, Ohno S, Murakami S, Hachitanda Y, Oda S: Prognostic value of epidermal growth factor receptor (EGFR) and its relationship to the estrogen receptor status in 1029 patients with breast cancer. Breast Cancer Res Treat. 2002, 71 (1): 67-75. 10.1023/A:1013397232011.CrossRefPubMed
38.
Zurück zum Zitat Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM: Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res. 2004, 10 (16): 5367-5374. 10.1158/1078-0432.CCR-04-0220.CrossRefPubMed Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C, Cowan D, Dressler L, Akslen LA, Ragaz J, Gown AM, Gilks CB, van de Rijn M, Perou CM: Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res. 2004, 10 (16): 5367-5374. 10.1158/1078-0432.CCR-04-0220.CrossRefPubMed
39.
Zurück zum Zitat Sternlicht MD, Sunnarborg SW: The ADAM17-amphiregulin-EGFR axis in mammary development and cancer. J Mammary Gland Biol Neoplasia. 2008, 13 (2): 181-194. 10.1007/s10911-008-9084-6.CrossRefPubMedPubMedCentral Sternlicht MD, Sunnarborg SW: The ADAM17-amphiregulin-EGFR axis in mammary development and cancer. J Mammary Gland Biol Neoplasia. 2008, 13 (2): 181-194. 10.1007/s10911-008-9084-6.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Walerych D, Napoli M, Collavin L, Del Sal G: The rebel angel: mutant p53 as the driving oncogene in breast cancer. Carcinogenesis. 2012, 33 (11): 2007-2017. 10.1093/carcin/bgs232.CrossRefPubMedPubMedCentral Walerych D, Napoli M, Collavin L, Del Sal G: The rebel angel: mutant p53 as the driving oncogene in breast cancer. Carcinogenesis. 2012, 33 (11): 2007-2017. 10.1093/carcin/bgs232.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Frazier DP, Kendig RD, Kai F, Maglic D, Sugiyama T, Morgan RL, Fry EA, Lagedrost SJ, Sui G, Inoue K: Dmp1 physically interacts with p53 and positively regulates p53′s stability, nuclear localization, and function. Cancer Res. 2012, 72 (7): 1740-1750. 10.1158/0008-5472.CAN-11-2410.CrossRefPubMedPubMedCentral Frazier DP, Kendig RD, Kai F, Maglic D, Sugiyama T, Morgan RL, Fry EA, Lagedrost SJ, Sui G, Inoue K: Dmp1 physically interacts with p53 and positively regulates p53′s stability, nuclear localization, and function. Cancer Res. 2012, 72 (7): 1740-1750. 10.1158/0008-5472.CAN-11-2410.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Sun Y, Rigas B: The thioredoxin system mediates redox-induced cell death in human colon cancer cells: implications for the mechanism of action of anticancer agents. Cancer Res. 2008, 68 (20): 8269-8277. 10.1158/0008-5472.CAN-08-2010.CrossRefPubMedPubMedCentral Sun Y, Rigas B: The thioredoxin system mediates redox-induced cell death in human colon cancer cells: implications for the mechanism of action of anticancer agents. Cancer Res. 2008, 68 (20): 8269-8277. 10.1158/0008-5472.CAN-08-2010.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Kim SJ, Miyoshi Y, Taguchi T, Tamaki Y, Nakamura H, Yodoi J, Kato K, Noguchi S: High thioredoxin expression is associated with resistance to docetaxel in primary breast cancer. Clin Cancer Res. 2005, 11 (23): 8425-8430. 10.1158/1078-0432.CCR-05-0449.CrossRefPubMed Kim SJ, Miyoshi Y, Taguchi T, Tamaki Y, Nakamura H, Yodoi J, Kato K, Noguchi S: High thioredoxin expression is associated with resistance to docetaxel in primary breast cancer. Clin Cancer Res. 2005, 11 (23): 8425-8430. 10.1158/1078-0432.CCR-05-0449.CrossRefPubMed
44.
Zurück zum Zitat Yamamoto M, Taguchi Y, Ito-Kureha T, Semba K, Yamaguchi N, Inoue J: NF-kappaB non-cell-autonomously regulates cancer stem cell populations in the basal-like breast cancer subtype. Nat Commun. 2013, 4: 2299-PubMed Yamamoto M, Taguchi Y, Ito-Kureha T, Semba K, Yamaguchi N, Inoue J: NF-kappaB non-cell-autonomously regulates cancer stem cell populations in the basal-like breast cancer subtype. Nat Commun. 2013, 4: 2299-PubMed
Metadaten
Titel
Phospho-aspirin (MDC-22) inhibits breast cancer in preclinical animal models: an effect mediated by EGFR inhibition, p53 acetylation and oxidative stress
verfasst von
Liqun Huang
Chi C Wong
Gerardo G Mackenzie
Yu Sun
Ka Wing Cheng
Kvetoslava Vrankova
Ninche Alston
Nengtai Ouyang
Basil Rigas
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2014
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-14-141

Weitere Artikel der Ausgabe 1/2014

BMC Cancer 1/2014 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.