You are on page 1of 17

14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023].

See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Received: 17 May 2019 Revised: 2 December 2019 Accepted: 22 December 2019
DOI: 10.1111/bph.14985

RESEARCH PAPER

Overcoming tamoxifen resistance in oestrogen receptor-


positive breast cancer using the novel thiosemicarbazone anti-
cancer agent, DpC

Sundus N. Maqbool1,2 | Syer C. Lim1 | Kyung Chan Park1 | Rumeza Hanif2 |


Des R. Richardson1 | Patric J. Jansson1 | Zaklina Kovacevic1

1
Molecular Pathology and Pharmacology
Program, Department of Pathology and Bosch Background and Purpose: Breast cancer is the leading cause of death in women
Institute, University of Sydney, Sydney, NSW,
worldwide, with resistance to current therapeutic strategies, including tamoxifen,
Australia
2
Atta-ur Rahman School of Applied causing major clinical challenges and leading to more aggressive and metastatic dis-
Biosciences, National University of Sciences ease. To address this, novel strategies that can inhibit the mechanisms responsible
and Technology, Islamabad, Pakistan
for tamoxifen resistance need to be assessed.
Correspondence Experimental Approach: We examined the effect of the novel, clinically-trialled,
Zaklina Kovacevic, Patric J. Jansson, and Des
R. Richardson, Molecular Pharmacology and thiosemicarbazone anti-cancer agent, DpC, and its potential as a combination therapy
Pathology Program, Department of Pathology with the clinically used estrogen receptor (ER) antagonist, tamoxifen, using both
and Bosch Institute, University of Sydney,
Sydney, NSW 2006, Australia. tamoxifen-resistant and -sensitive, human breast cancer cells (MDA-MB-453, MDA-
Email: zaklina.kovacevic@sydney.edu.au; MB-231 and MCF-7) in 2D and 3D cell-culture. Synergy was assessed using the
patric.jansson@sydney.edu.au; d.richardson@
med.usyd.edu.au Chou-Talalay method. The molecular and anti-proliferative effects of these agents
and their combination was examined via Western blot, immunofluorescence and col-
Funding information
Cancer Institute New South Wales, Grant/ ony formation assays.
Award Numbers: CDF141147 and Key Results: Combinations of tamoxifen with DpC were highly synergistic, leading to
CDF171126; National Health and Medical
Research Council, Grant/Award Numbers: potent inhibition of cell proliferation, colony formation, and ER-α transcriptional
1062607, 1159596 and APP1140447; activity. The combination also more efficiently reduced major molecular drivers of
National Breast Cancer Foundation, Grant/
Award Number: 1146599; Cancer Australia; proliferation of tamoxifen-resistant cells, including c-Myc, cyclin D1, and p-AKT,
Cure Cancer Australia Foundation, Grant/ while up-regulating the cell cycle inhibitor, p27, and inhibiting oncogenic phosphory-
Award Number: 1086449
lation of ER-α at Ser167. Assessing these effects using 3D cell culture further con-
firmed the greater effects of DpC combined with tamoxifen in reducing ER-α
expression, and that of the proliferation marker, Ki-67, in both tamoxifen-sensitive
and -resistant MCF-7 spheroids.
Conclusions and Implications: These studies demonstrate that the synergistic combi-
nation of DpC with tamoxifen could be a promising new therapeutic strategy to over-
come tamoxifen resistance in ER-positive breast cancer.

Abbreviations: 4-OHT, 4-hydroxy-tamoxifen; CI, combination index; DpC, di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone; EGFR, EGF receptor; ER, oestrogen receptor; EREs,
oestrogen response elements; IGF1R, insulin-like growth factor 1 receptor.

Br J Pharmacol. 2020;177:2365–2380. wileyonlinelibrary.com/journal/bph © 2020 The British Pharmacological Society 2365


14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2366 MAQBOOL ET AL.

1 | I N T RO DU CT I O N
What is already known
Breast cancer is a leading cause of cancer-related deaths, affecting 2.1
• Tamoxifen resistance is a major clinical problem, leading
million women each year (Ali et al., 2016). The oestrogen receptor-α
to development of aggressive, metastatic breast cancers.
(ER-α) is expressed in 75% of breast cancer cases (Zhang, Man, Zhao,
• Intrinsic and ligand-independent mechanisms of ER-α
Dong, & Ma, 2014) and is a ligand-responsive transcription factor that
activation drive development of tamoxifen resistance.
plays a pivotal role in driving breast cancer progression (Hayashi et al.,
2003). The orthodox genomic ER-α pathway entails oestradiol (E2)
What this study adds
binding to ER-α, resulting in dimerization and nuclear localization,
• The novel clinically trialled anti-cancer agent DpC inhibits
where it binds to oestrogen response elements (EREs) to promote
major pathways that drive ligand-independent ER-α
transcription responsible for growth, proliferation, and metastasis
activation.
(Acconcia & Kumar, 2006; Klinge, 2001).
• Combining DpC with tamoxifen demonstrates potent
Therapies for ER-positive breast cancer include selective ER
synergy and overcomes tamoxifen resistance in breast
modulators, with tamoxifen being the standard of care for younger
cancer.
women with breast cancer (Osborne, 1998). Tamoxifen binds ER-α,
blocking its ability to bind EREs (Obrero, Yu, & Shapiro, 2002). How-
What is the clinical significance
ever, 20% of all ER-positive breast cancer develop resistance to
• Combining DpC with tamoxifen may overcome tamoxifen
tamoxifen, leading to more aggressive neoplasms (Ali et al., 2016;
resistance, preventing onset of more aggressive breast
Ring & Dowsett, 2004). Several mechanisms drive tamoxifen resis-
cancer.
tance, including the interaction of ER-α with growth factors, such as
• These findings are highly significant, as 20% of ER-
EGF and/or the over-expression of human EGF receptors (i.e., EGFR
positive breast cancer patients develop tamoxifen
and HER2), activating downstream MAPK and PI3K/AKT pathways
resistance.
(Campbell et al., 2001; Fan, Wang, Santen, & Yue, 2007; Ghayad
et al., 2010; Osborne, Shou, Massarweh, & Schiff, 2005; Riggins,
Schrecengost, Guerrero, & Bouton, 2007). This enables ligand-
independent phosphorylation of ER-α at Ser118 and Ser167
(Campbell et al., 2001; Ghayad et al., 2010; Kato et al., 1995; Sun Considering this, we examined the role of DpC and its potential
et al., 2001), promoting c-Myc and cyclin D1 expression (Ali et al., for synergy with tamoxifen in ER-positive breast cancer. We have
2016; Green et al., 2016; Kilker, Hartl, Rutherford, & Planas-Silva, demonstrated that DpC combined with the active tamoxifen metabo-
2004; Santen et al., 2009). In fact, over-expression or activation of lite, 4-hydroxy-tamoxifen (4-OHT), down-regulates key oncogenes,
these latter proteins is extensively linked to tamoxifen resistance including p-AKT, c-Myc, and cyclin D1, while potently inhibiting prolif-
(Santen et al., 2009), and it is critical to develop novel strategies to eration and colony formation of tamoxifen-resistant and -sensitive
inhibit or prevent resistance. MCF-7 cells. Hence, the combination of DpC with tamoxifen has the
We synthesized the novel thiosemicarbazone, di-2-pyridylketone potential to overcome tamoxifen resistance and prevent the progres-
4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), which has potent sion of breast cancer.
and selective anti-cancer activity against multiple neoplasms in vitro
and in vivo (Guo et al., 2016; Kovacevic, Chikhani, Lovejoy, & Richard-
son, 2011; Lovejoy et al., 2012). DpC binds iron and copper to form 2 | METHODS
redox-active complexes that are cytotoxic to cancer cells (Lane,
Saletta, Suryo Rahmanto, Kovacevic, & Richardson, 2013) and up- 2.1 | Cell culture
regulates the metastasis suppressor, N-myc downstream regulated
gene-1 (Kovacevic et al., 2011). Following extensive pharmacokinetic The human breast cancer cell lines, MCF-7 (RRID:CVCL_0031),
and toxicity studies (Kovacevic et al., 2011; Lovejoy et al., 2012; T47D (RRID:CVCL_0553), MDA-MB-453 (RRID:CVCL_0418), and
Sestak et al., 2015), DpC entered Phase I clinical trials to assess its MDA-MB-231 (RRID:CVCL_0062), were purchased from the Ameri-
efficacy in advanced cancers (Jansson et al., 2015). can Type Culture Collection (Rockville, MD). Cells were grown in
Importantly, DpC decreases the expression of key EGF receptors MEM complete media (ThermoFisher Scientific, MA, USA) sup-
including EGFR, HER2, and HER3 (Kovacevic et al., 2011), while plemented with 10% FCS (ThermoFisher Scientific). Tamoxifen-
inhibiting downstream PI3K/AKT and MAPK signalling (Kovacevic resistant MCF-7 cells (resistant) and their sensitive vehicle control
et al., 2011; Kovacevic et al., 2016; Lui et al., 2015; Xi et al., 2017). As counterparts (sensitive) were generated (Knowlden et al., 2003)
these pathways play major roles in tamoxifen resistance (Butt, McNeil, and generously provided by Dr. Elizabeth Caldon (Garvan Institute
Musgrove, & Sutherland, 2005; Ghayad et al., 2010; Kato et al., 1995; of Medical Research, Sydney). The resistant cells were generated
Riggins et al., 2007; Sun et al., 2001), DpC may overcome this major by continuous incubation with the active metabolite of tamoxifen,
clinical problem. namely, 4-OHT, for 6 months (Knowlden et al., 2003). All cells
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAQBOOL ET AL. 2367

were authenticated based on viability, recovery, growth, morphol- their combination (5 μM each) for 24 hr/37 C, cells were then
ogy, and cytogenetic analysis (i.e., antigen expression, DNA profile, harvested and protein extracted using standard methods (Chen et al.,
and iso-enzymology) by the provider. 2012). Western blot analysis was performed via established methods
The cell lines were maintained in phenol red-free RPMI media (Gao & Richardson, 2001). The primary antibodies used (diluted at
(ThermoFisher Scientific), supplemented with 5% (v/v) charcoal- 1:1,000) included ER-α (RRID:AB_2632959), p-ER-α (Ser167; RRID:
stripped serum (csFBS; Sigma-Aldrich; MO, USA) and 100 μgml−1 AB_2102074), EGFR (RRID:AB_2262057), p-EGFR (Tyr1068; RRID:
penicillin/streptomycin/glutamine (ThermoFisher Scientific). Resistant AB_1903957), AKT (RRID:AB_329827), p-AKT (Ser473; RRID:
cells were grown in 10 μM 4-OHT (re-suspended in tetrahydrofuran) AB_329824), c-Myc (RRID:AB_2151827), p-c-Myc (Ser62; RRID:
along with 10 pM β-E2 (re-suspended in ethanol). Sensitive cells were AB_2151830), cyclin D1 (RRID:AB_2228523), p27 (RRID:
maintained in base media with 10 μM of tetrahydrofuran and 10 pM AB_10693314), SRC3 (RRID:AB_823642), NF-κB-p65 (RRID:
of ethanol. Cells were grown in an incubator at 37 C in a humidified AB_2341215), and p-NF-κB-p65 (Ser536; RRID:AB_331281; Cell Sig-
atmosphere of 5% CO2 and 95% air. For studies assessing the effects naling Technology, MA); β-actin (diluted at 1:10,000, RRID:
of E2 and EGF, cells were incubated with phenol-red free media con- AB_476692) was purchased from Sigma-Aldrich. The secondary anti-
taining 5% csFBC for 3 days prior to treatment with E2 and EGF at bodies implemented (diluted 1:10,000) include anti-rabbit and anti-
−1
concentrations of 10 nM and 100 ngml , respectively, for mouse antibodies (Sigma-Aldrich; RRID:AB_2336059, RRID:
10 min/37 C. AB_2336060).

2.2 | MTT assay 2.5 | Spheroid generation and


immunofluorescence
Both tamoxifen-sensitive and -resistant MCF-7 cells were seeded in
96-well plates and allowed to attach overnight. Cells were then Spheroids were prepared by seeding 35,000 cells per well in phenol
treated with DpC or 4-OHT at a range of concentrations (0–1.25 μM red-free RPMI supplemented with 5% csFBS and 100 μgml−1
for DpC and 0–40 μM for 4-OHT) and incubated for a further 72 hr. penicillin/streptomycin/glutamine in 96-well plates coated with
Proliferation was examined using the 3-(4,5-dimethylthiazol-2-yl)- 0.75% (w/v) agarose. The plates were then incubated for
2,5-diphenyl tetrazolium (MTT; Sigma-Aldrich) assay, using standard 3–4 days/37 C in 5% CO2. Once established, spheroids were treated
methods (Richardson, Tran, & Ponka, 1995). As shown previously, with either 5-μM DpC, 5-μM 4-OHT, or these agents combined at
MTT colour formation was directly proportional to viable cell number 5 μM each (combination) for a further 24 hr/37 C. Spheroids were
(Richardson et al., 1995). then fixed in 4% paraformaldehyde for 3 hr/20 C. Following fixation,
spheroids were washed three times with PBS, permeabilized with
0.1% Triton X-100, and blocked using 5% BSA in PBS overnight at
2.3 | Chou–Talalay combination analysis 4 C. They were then incubated with either mouse Ki-67 (RRID:
AB_2797703) or rabbit ER-α monoclonal antibodies (Cell Signaling
For the combination studies with DpC and 4-OHT, the Chou–Talalay Technology) overnight at 4 C. After washing 3 × 10 min with PBS,
method was used (Chou, 2006; Chou, 2010). The resulting combina- secondary antibodies (anti-mouse IgG Alexa Fluor 488 [RRID:
tion index (CI) offers a quantitative definition for an additive effect, AB_10694704] and anti-rabbit IgG Alexa Fluor 594 [RRID:
where synergism is evident when CI < 1 and antagonism when CI > 1 AB_2716249]; Cell Signaling Technology) were added at 1:200 dilu-
in drug combinations (Chou, 2010). Briefly, to calculate the CI and tion in BSA buffer for 1 hr/20 C. Nuclei were stained with DAPI (Life
dose reduction index, cells were incubated with DpC and 4-OHT in a Technologies), and spheroids imaged and photographed using a META
range that was eight times below, and eight times above, their respec- confocal microscope (Carl Zeiss, Germany). Intensity analysis was per-
tive IC50 values after a 72 hr/37 C incubation (i.e., 0.125, 0.25, 0.5, formed using Fiji ImageJ software (NIH, Baltimore, MD; RRID:
1, 2, 4, and 8 × IC50). The concentrations of DpC and 4-OHT used SCR_003070).
were cell type dependent, as each cell line had its own IC50 values for
these agents. The resulting dose–response curves were then analysed
using Chou–Talalay methodology (Chou, 2006; Chou, 2010). 2.6 | Colony formation assay

MCF-7-sensitive and -resistant cells were harvested and plated


2.4 | Protein extraction, Western blot, and (70 cells per plate) in 100-mm Petri dishes for 21 days with media
antibodies containing either DpC (0.125 μM), 4-OHT (0.125 μM), or their combi-
nation at 0.125 μM each. Colonies were then rinsed with 10-ml PBS,
The antibody-based procedures used in this study comply with the fixed (acetic acid/methanol [1:7 v/v]), and stained with 0.5% crystal
recommendations made by the British Journal of Pharmacology. Fol- violet. After staining, colonies were visualized and examined
lowing treatment with either control, DpC (5 μM), 4-OHT (5 μM), or microscopically.
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2368 MAQBOOL ET AL.

2.7 | Dual-luciferase assay for ER-α 3 | RE SU LT S

The ERE Luciferase Reporter Assay Kit (Cignal Reporter Assay Kit; 3.1 | Assessing the synergy between DpC and
Qiagen, Netherlands) was used following the manufacturer's proto- 4-OHT in breast cancer cells
col. Briefly, the MCF-7-sensitive and -resistant cells were trans-
fected with the ERE Luciferase Reporter, as well as the relevant The novel anti-cancer agent, DpC, and its potential to synergize
positive and negative controls for 48 hr. Cells were then incubated with the active metabolite of tamoxifen, 4-OHT, was
with either DpC (5 μM), 4-OHT (5 μM), or their combination (5 μM assessed using well-characterized ER-a positive (MCF-7 and T47D)
each), in the presence of E2 for a further 24 hr. Luciferase activity and ER-a negative (MDA-MB-231 and MDA-MB-453) breast
was then assessed implementing the Dual-Luciferase Reporter cancer cells (Miller & Katzenellenbogen, 1983; Vranic, Gatalica, &
Assay kit (Cat. No. 1910; Promega) using a luminometer (FLUOstar; Wang, 2011).
BMG Labtech, Germany). DpC substantially inhibited proliferation of all breast cancer cells
examined, with IC50 values (72 hr) ranging from 0.006 to 0.024 μM
(Figure 1a). ER-positive breast cancer cells (T47D and MCF-7) were
2.8 | Densitometry most sensitive, while triple-negative MDA-MB-231 cells were least
sensitive to DpC (Figure 1a). 4-OHT alone displayed much higher IC50
Densitometry was performed using Quantity One software (Bio-Rad; values than DpC, ranging from 7 to 14 μM (72 hr; Figure 1b). As
CA; RRID:SCR_014280) and normalized using the β-actin loading expected, ER-positive cells were two to three times more sensitive to
control. 4-OHT compared to ER-negative cells (MDA-MB-453 and MDA-MB-
231; Figure 1b).
Next, we examined the potential synergy between DpC and
2.9 | Data and statistical analysis 4-OHT using the Chou–Talalay method (Chou, 2010). For ER-
positive MCF-7 and T47D cells, the combination of DpC with
Data are presented as mean ± SD. The group data subjected to 4-OHT was highly synergistic (Figure 1c). In contrast, the ER-
statistical analysis have a minimum of n = 5 independent samples negative MDA-MB-231 and MDA-MB-453 cells had CI values close
per group. Statistical comparisons between two groups were evalu- to or >1, indicating no synergism (Figure 1c). These results suggest
ated by the unpaired Student's t test and were considered signifi- that the combination of DpC and 4-OHT was synergistic only in ER-
cant when P < .05. The data and statistical analysis comply with positive breast cancer cells.
the recommendations of the British Journal of Pharmacology on
experimental design and analysis in pharmacology (Curtis et al.,
2018). To provide blinding in our experiments, analysis of the 3.2 | Assessing synergy between DpC and 4-OHT
experimental data was performed by different people to those that in tamoxifen-sensitive and -resistant cells
completed the experiment. All studies were designed to provide
randomization, with cells being randomly assigned to different We next investigated the potential of combining DpC with 4-OHT in
treatment groups. overcoming tamoxifen resistance in ER-positive cells using
tamoxifen-resistant MCF-7 (resistant) cells and their sensitive coun-
terparts (sensitive; Knowlden et al., 2003). MTT studies were con-
2.10 | Materials ducted to assess the IC50 values of each agent in these cells under
different conditions, including (a) no ligands, (b) E2 alone (10 nM),
DpC was synthesized as described previously (Lovejoy et al., 2012). (c) EGF alone (100 ngml−1), or (d) E2 (10 nM) and EGF (100 ngml−1)
4-OHT was supplied by Sigma-Aldrich (MO, USA) and EGF was sup- together, for 72 hr. This was done as E2 or EGF can activate ER-α,
plied by Cell Signaling Technology (MA, USA). with EGF also promoting tamoxifen resistance (Ghayad et al., 2010;
Knowlden et al., 2003).
Tamoxifen-sensitive MCF-7 cells were highly sensitive to DpC
2.11 | Nomenclature of targets and ligands under all conditions assessed, with IC50 values <0.02 μM
(Figure 1d). Tamoxifen-resistant cells had significantly higher IC50
Key protein targets and ligands in this article are hyperlinked to values for DpC in the absence of ligands, although sensitivity was
corresponding entries in http://www.guidetopharmacology.org, the restored upon addition of E2, EGF, or their combination
common portal for data from the IUPHAR/BPS Guide to PHARMA- (Figure 1d). The IC50 values for 4-OHT in the sensitive cells
COLOGY (RRID:SCR_013077; Harding et al., 2018), and are perma- (Figure 1e) were up to 100-fold higher compared to DpC
nently archived in the Concise Guide to PHARMACOLOGY (Figure 1d). As expected, the IC50 values for 4-OHT in the resistant
2019/20 (Alexander, Cidlowski et al., 2019; Alexander, Fabbro cells were significantly higher, or slightly higher (i.e., for E2), when
et al., 2019a, 2019b). compared to their sensitive counterparts (Figure 1e). However, in
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAQBOOL ET AL. 2369

F I G U R E 1 The combination of DpC and 4-OHT is synergistic in ER-positive breast cancer cells. IC50 values for (a) DpC and (b) 4-OHT
in MCF-7, T47D, MDA-MB-231, and MDA-MB-453 breast cancer cells. (c) Combination index (CI) values of DpC in combination with
4-OHT in MCF-7, T47D, MDA-MB-231, and MDA-MB-453 breast cancer cells. IC50 values for (d) DpC and (e) 4-OHT in MCF-7-sensitive
and -resistant cells in the presence of control media or media containing E2, EGF, or both. (f) CI values of DpC combined with 4-OHT in
MCF-7-sensitive and -resistant cells in control media or media containing E2, EGF, or both. Data represent the mean ± SD of five
independent experiments. *P < .05, significantly different from the respective control; #P < .05, significantly different from the
corresponding treatment in MCF-7-sensitive cells
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2370 MAQBOOL ET AL.

the presence of E2, EGF, or their combination, the resistant using different doses of DpC and 4-OHT (Tables S1 and S2) further
cells also became significantly more sensitive to 4-OHT compared confirmed that these agents were synergistic in the majority of combi-
with the control (Figure 1e). nations. Further, dose reduction index values (Tables S1 and S2) indi-
For both sensitive and resistant cells, a synergistic effect was cated the doses of DpC and 4-OHT could be reduced to maintain
observed when DpC and 4-OHT were combined at the same dose efficacy, while potentially reducing toxicity, which would be highly
ratios, with CI values <1 under all conditions (Figure 1f). Notably, beneficial in clinical use. Overall, these studies demonstrate that DpC
synergy was more pronounced in control and E2-treated resistant cells in combination with 4-OHT exhibited a synergistic effect in ER-
when compared with their sensitive counterparts. Assessing CI values α-positive cell types.

F I G U R E 2 The effect of DpC, 4-OHT, and their combination on ER-α levels and transcriptional activity. Western blot analysis of total
ER-α and its phosphorylation at Ser167 in (a) sensitive and (b) tamoxifen-resistant MCF-7 cells treated with either control media or media
containing DpC (5 μM), 4-OHT (5 μM), or their combination (5 μM each) in the presence or absence of E2. β-actin was used as a protein
loading control. Data represent the mean ± SD of five independent experiments. *P < .05, significantly different from the respective
controls. Luciferase activity of ERE-containing constructs in MCF-7 (c) sensitive and (d) resistant cells treated with either control media or
media containing DpC, 4-OHT, or their combination. Data represent the mean ± SD of five independent experiments. *P < .05, significantly
different from the respective control
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAQBOOL ET AL. 2371

3.3 | DpC alone or in combination with 4-OHT p65 (Gionet, Jansson, Mader, & Pratt, 2009; Osborne et al., 2003).
decreases ER-α expression SRC3 was reduced by DpC and the combination in sensitive cells,
while being reduced by DpC, 4-OHT, and their combination in resis-
To elucidate the mechanisms underlying the synergy between DpC tant cells (Figure S1). While total NF-κB-p65 was not affected by
and 4-OHT, we next examined the effect of DpC alone or in combina- these agents in sensitive cells, DpC, 4-OHT, and their combination
tion with 4-OHT on ER-α in tamoxifen-sensitive and -resistant MCF-7 reduced NF-κB-p65 in resistant cells. Notably, in the presence of E2,
cells (Figure 2a,b). Both cell types were incubated for 24 hr with either the combination treatment was most effective at reducing SRC3 and
DpC (5 μM), 4-OHT (5 μM), or these agents combined (5 μM each), NF-κB-p65 levels in the resistant cells (Figure S1), demonstrating the
followed by a 10-min incubation with or without the ER-α activating synergy between DpC and 4-OHT. The activating phosphorylation of
ligand, E2 (10 nM). These doses were selected based on our studies NF-κB-p65 (Ser536) was also most reduced by the combination in
using DpC in vitro (Kovacevic et al., 2011) and studies by others using sensitive cells. However, its expression in the resistant cells was
4-OHT (Lee et al., 2018). The levels of total ER-α or its phosphoryla- increased by DpC or 4-OHT (Figure S1), which may account for the
tion at Ser167 (p-ER-α), which contributes to tamoxifen resistance lower sensitivity of these cells to these agents alone (Figure 1d,e).
(Bostner et al., 2013; Campbell et al., 2001), were then assessed via Importantly, this adverse effect was prevented when resistant cells
Western blot. were incubated with DpC and 4-OHT as a combination (Figure S1),
In sensitive cells, DpC significantly down-regulated ER-α and p- further highlighting their synergy.
ER-α (Figure 2a), while 4-OHT markedly increased ER-α and p-ER-α
under all conditions (Figure 2a). This was expected, as 4-OHT binds to
ER-α, leading to its stabilization and the inhibition of downstream sig- 3.5 | DpC down-regulates AKT activation in
nalling (Jaiyesimi, Buzdar, Decker, & Hortobagyi, 1995). Assessing the tamoxifen-sensitive and -resistant MCF-7 cells
ratio of p-ER-α to total ER-α for 4-OHT demonstrated a significant
increase, compared with the control, suggesting that 4-OHT facili- To determine whether the synergy between DpC and 4-OHT was due
tated ER-α phosphorylation (Figure 2a). In contrast, this ratio did not to inhibition of EGFR signalling, we assessed total EGFR and its acti-
significantly alter for DpC relative to the control, suggesting that a vating phosphorylation at Tyr1068 in tamoxifen-sensitive and
decrease in ER-α phosphorylation can be attributed to decreased total -resistant MCF-7 cells.
ER-α. When DpC was combined with 4-OHT (combination), total ER- Considering its ability to down-regulate EGFR in other cell types
α and p-ER-α remained similar to the untreated controls under all (Kovacevic et al., 2016), it was unexpected that DpC alone or in com-
conditions. bination with 4-OHT up-regulated EGFR in sensitive and resistant
In resistant cells, DpC again markedly reduced total ER-α, while cells, regardless of E2 treatment (Figure 3a,b). Further, DpC and the
4-OHT had no significant effect (Figure 2b). In contrast to sensitive combination of DpC with 4-OHT also distinctly up-regulated p-EGFR,
cells (Figure 2a), DpC significantly increased p-ER-α in the absence of although this was only evident in the presence of E2 in sensitive cells
E2, while reducing its levels in the presence of E2 in resistant cells (Figure 3a). In resistant cells, DpC also significantly increased p-EGFR
(Figure 2b). The combination of DpC with 4-OHT in resistant cells when E2 was present (Figure 3b), although this effect was far less pro-
gave similar results to DpC alone (Figure 2b). nounced than in sensitive cells (Figure 3a).
Conversely, 4-OHT had no significant effect on total or phos-
phorylated EGFR in the sensitive cells (Figure 3a), while a slight but
3.4 | DpC and 4-OHT inhibit ER-α transcriptional significant reduction in total EGFR was observed in resistant cells in
activity in sensitive and resistant MCF-7 cells the absence of E2 (Figure 3b). The ratio of p-EGFR/EGFR was signifi-
cantly increased for DpC and the combination in sensitive cells in
Further studies assessed ER-α transcriptional activity, as activated ER- the presence of E2 (Figure 3a), while a significant decrease in this
α can directly or indirectly bind to target gene EREs to promote breast ratio was observed under these conditions in resistant cells
cancer progression (Levin & Pietras, 2008). To achieve this, ERE dual- (Figure 3b). Overall, these results suggest that DpC promotes EGFR
luciferase reporter assays were performed, which was transfected into phosphorylation in sensitive MCF-7 cells to a much greater extent
tamoxifen-sensitive and -resistant MCF-7 cells followed by incubation than in resistant cells.
with DpC (5 μM), 4-OHT (5 μM), or their combination for 24 hr. To further investigate this, we examined these cells in the pres-
A significant decrease in luciferase activity was observed in sensi- ence of E2 and EGF together, as addition of EGF results in more
tive (Figure 2c) and resistant (Figure 2d) cells upon treatment with robust EGFR activation (Stabile et al., 2005). Under control conditions,
DpC or 4-OHT. The combination of DpC and 4-OHT also led to a pro- endogenous EGFR was very low in sensitive cells, with E2 + EGF hav-
nounced down-regulation of luciferase activity in sensitive and resis- ing no substantial effect on EGFR (Figure S2A). In response to DpC
tant cells, with the combination being most effective in resistant cells alone, total and p-EGFR levels were markedly increased in sensitive
(Figure 2d). cells (Figure S2A), as observed with E2 alone (Figure 3a). However, the
Considering reduced ER-α transcriptional activity, we further stimulatory effect of EGF + E2 on p-EGFR was inhibited when DpC
examined two major co-activators of ER-α, namely, SRC3 and NF-κB- was combined with 4-OHT (Figure S2A).
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2372 MAQBOOL ET AL.

F I G U R E 3 DpC inhibits downstream AKT activation in tamoxifen-sensitive and -resistant cells. Western blot analysis of total EGFR and its
phosphorylation at Tyr1068, along with total AKT and its phosphorylation at Ser473 in (a) tamoxifen-sensitive and (b) tamoxifen-resistant MCF-7
cells treated with either control media or media containing DpC (5 μM), 4-OHT (5 μM), or their combination (5 μM each) in the presence or
absence of E2. β-actin was used as a protein loading control. Data represent the mean ± SD of five independent experiments. *P < .05,
significantly different from the respective control

The resistant cells had much higher endogenous EGFR and to (Figure S2A). However, this increase of p-EGFR in resistant cells
a lesser extent, p-EGFR, than sensitive cells (Figure S2A). Further, incubated with E2 and EGF was not observed with the combina-
resistant cells responded strongly to E2 + EGF treatment, with a tion treatment (Figure S2A). Hence, in the presence of E2 + EGF,
pronounced increase in p-EGFR observed after control, DpC, and the DpC and 4-OHT combination potently inhibited EGFR
4-OHT treatment, compared with the control without E2 + EGF activation.
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAQBOOL ET AL. 2373

To determine whether other major receptors were affected by markedly affected by DpC or 4-OHT in sensitive cells, this receptor
these chemotherapeutic agents, we also assessed HER2 and insulin- was reduced by DpC and the combination in resistant cells, regardless
like growth factor 1 receptor (IGF1R) in sensitive and resistant cells, of E2 (Figure S2B). Further, the combination of DpC with 4-OHT
as these receptors contribute to tamoxifen resistance (Osborne et al., decreased IGF1R levels in sensitive cells, while having no distinct
2003; Osborne et al., 2005; Riggins et al., 2007). While HER2 was not effect in resistant cells (Figure S2B).

F I G U R E 4 Combining DpC with 4-OHT inhibits c-Myc and cyclin D1, while promoting p27 expression. Western blot analysis of c-Myc and
its phosphorylation site at Ser62, along with cyclin D1 and p27 in (a) sensitive- and (b) tamoxifen-resistant MCF-7 cells treated with either control
media or media containing DpC (5 μM), 4-OHT (5 μM), or their combination (5 μM each) in the presence or absence of E2. β-actin was used as a
protein loading control. Data represent the mean ± SD of five independent experiments. *P < .05, significantly different from the respective
control
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2374 MAQBOOL ET AL.

As EGFR, HER2, and IGF1R signalling pathways converge on sim- and significantly increasing the cyclin-dependent kinase inhibitor, p27
ilar downstream targets, we investigated their major target, AKT, and (Figure 4b).
its activating phosphorylation at Ser473 (Levin & Pietras, 2008), in While the above studies were conducted with DpC at 5 μM,
sensitive and resistant MCF-7 cells (Figure 3a,b). Once activated via lower doses of DpC (i.e., 0.1, 0.5, and 1 μM) alone or combined with
phosphorylation, AKT directly phosphorylates ER-α at Ser167, which 5-μM 4-OHT induced similar, dose-dependent effects on ER-α, p-ER-
is a major contributor to tamoxifen resistance (Campbell et al., 2001). α, p-AKT, p-c-Myc, and cyclin D1 in both sensitive and resistant cells
In sensitive cells, total AKT was significantly reduced by the combina- (Figure S3).
tion in the absence of E2, while no distinct effect was observed under
other conditions (Figure 3a). Interestingly, 4-OHT markedly increased
p-AKT in the absence of E2, while p-AKT was significantly reduced by 3.7 | Combining DpC with 4-OHT decreases
DpC alone or the combination in the presence of E2 (Figure 3a). A proliferation and inhibits colony formation
similar effect was observed in resistant cells, with DpC and the combi-
nation also significantly decreasing p-AKT under all conditions To further assess the synergy between DpC and 4-OHT using more
(Figure 3b). pathologically relevant tumour models, we examined these agents
Taken together, our results showed that AKT activation was using 3D cell culture, where MCF-7-sensitive and -resistant cells were
substantially inhibited in resistant cells, suggesting a mechanism grown as spheroids. Spheroids better represent the in vivo setting and
by which DpC and its combination with 4-OHT overcome are more resistant to chemotherapy (Edmondson, Broglie, Adcock, &
resistance. Yang, 2014; Nyga, Cheema, & Loizidou, 2011). Once established,
spheroids were treated with DpC (5 μM), 4-OHT (5 μM), or their com-
bination for 24 hr and examined for ER-α and Ki-67 (marker of prolif-
3.6 | DpC combined with 4-OHT attenuates c-Myc eration; Yerushalmi, Woods, Ravdin, Hayes, & Gelmon, 2010) via
and cyclin D1, while promoting p27 expression immunofluorescence.
A significant down-regulation of ER-α and Ki-67 was observed
We next assessed the major downstream targets of ER-α and AKT, in sensitive and resistant spheroids treated with DpC alone or in
including c-Myc and cyclin D1, both of which play oncogenic roles in combination with 4-OHT (Figure 5a). Notably, 4-OHT alone did
breast cancer and are associated with tamoxifen resistance (Butt not affect ER-α and Ki-67 under these conditions. These results
et al., 2005; Green et al., 2016; Sears, 2004). We also examined the support our current findings that down-regulation of c-Myc and
tumour suppressor, p27, a major cyclin D1 inhibitor that attenuates cyclin D1 by DpC and the combination decreases proliferation. We
proliferation (Chiarle, Pagano, & Inghirami, 2001). also performed colony formation assays to determine the ability of
In sensitive cells, c-Myc was significantly down-regulated under sensitive and resistant cells to undergo unlimited division. While
all conditions, with the combination having the most potent effect control cells formed large colonies, no colony formation was
(Figure 4a). In these cells, c-Myc was detected as two bands (~50 and observed with DpC or the combination (Figure 5b,c). Further,
60 kDa), which may reflect different isoforms (Benassayag et al., 4-OHT reduced colony formation in sensitive cells, but not in
2005), or post-translational modification (Hann, 2006), with quantita- resistant cells where colonies were similar to the control
tion assessing both bands as a total. Next, we examined phosphory- (Figure 5b,c), which further confirms that these cells are resistant
lated c-Myc (Ser62), as this phosphorylation is associated with to tamoxifen.
increased c-Myc stability and oncogenic activity (Sears, 2004; Yeh
et al., 2004). Both DpC and the combination significantly decreased p-
c-Myc under all conditions (Figure 4a). 4-OHT also potently reduced 4 | DI SCU SSION
p-c-Myc, but only in the presence of E2 in sensitive cells. A similar
trend was observed for cyclin D1, with DpC and the combination sig- Various pathways contribute to tamoxifen resistance, with ER-α over-
nificantly decreasing cyclin D1 under all conditions (Figure 4a). p27 expression and its crosstalk with multiple oncogenic pathways such as
was significantly increased by 4-OHT in sensitive cells, while DpC had EGFR and its downstream targets (i.e., AKT, c-Myc, and cyclin D1)
no significant effect (Figure 4a). being major players (Butt et al., 2005; Campbell et al., 2001; Chang,
In resistant cells, c-Myc was predominantly expressed as the 2012; Fan et al., 2007; Ghayad et al., 2010; Knowlden et al., 2003).
upper band (Figure 4b). DpC alone again significantly reduced total c- We demonstrated that the novel, clinically tested anti-cancer agent,
Myc, p-c-Myc, and cyclin D1, while having no effect on p27 DpC, inhibits multiple oncogenic pathways, including EGFR, AKT, c-
(Figure 4b). However, in direct contrast to its effects in sensitive cells Myc, and cyclin D1, in pancreatic and prostate cancer cells (Kovacevic
(Figure 4a), 4-OHT failed to decrease total c-Myc or cyclin D1, while et al., 2011; Kovacevic et al., 2016; Xi et al., 2017), leading to potent
significantly reducing p27 in resistant cells (Figure 4b). This is in agree- anti-tumour activity in vitro and in vivo (Kovacevic et al., 2011;
ment with these cells being tamoxifen resistant (Kilker & Planas-Silva, Lovejoy et al., 2012). These studies prompted us to examine the effi-
2006). The combination was most effective in resistant cells, signifi- cacy of DpC against breast cancer and its potential to overcome
cantly decreasing c-Myc phosphorylation and cyclin D1 expression tamoxifen resistance.
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAQBOOL ET AL. 2375

F I G U R E 5 DpC and 4-OHT inhibit 3D growth and colony formation of tamoxifen-resistant and -sensitive MCF-7 cells. (a) Representative
images of ER-α (red), Ki-67 (green), and DAPI (blue) staining in multicellular 3D tumour spheroids of MCF-7-sensitive and -resistant cells treated
with either control, DpC (5 μM), 4-OHT (5 μM), or their combination (5 μM each) for 24 hr. The scale bar in the bottom right-hand corner of the
first image in each set represents 2 μm and is the same across all images. Quantification of pixel intensity for ER-α and Ki-67 was performed
using ImageJ software and expressed as mean ± SD (five experiments). *P < .05, significantly different from the respective control. (b) Colony
formation was analysed in tamoxifen-sensitive and -resistant MCF-7 cells in the presence of control media or media containing DpC (0.125 μM),
4-OHT (0.125 μM), or their combination (0.125 μM each) for 21 days. Higher magnification (10×) images are shown in (c). Scale bar in the bottom
right-hand corner of the first image in (c) represents 2 μm and is the same across all images. Data represent five independent experiments
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2376 MAQBOOL ET AL.

Here, we have demonstrated that DpC has potent anti- major ER-α co-activators, namely, SRC3 and NF-κB-p65, further con-
proliferative activity against ER-positive, HER2 over-expressing, and tributing to the reduced ER-α transcriptional activity. In fact, NF-κB-
triple-negative breast cancer cells. DpC was least effective against p65 and SRC3 both contribute to tamoxifen resistance in breast can-
MDA-MB-231 triple-negative cells, and this may be due to the cer, and their inhibition may be a vital aspect of the synergy between
absence of both ER-α and HER2 (Holliday & Speirs, 2011), which DpC and 4-OHT in resistant cells (Biswas, Singh, Shi, Pardee, &
we demonstrate are major DpC targets. DpC was most effective Iglehart, 2005; Osborne et al., 2003).
against ER-positive cells, potently inhibiting proliferation and colony The effect of DpC on reducing cyclin D1 and c-Myc may further
formation for tamoxifen-sensitive and -resistant MCF-7 cells. Fur- contribute to the synergy between DpC and 4-OHT, as cyclin D1 and
ther, Chou–Talalay analysis demonstrated that the combination of c-Myc are necessary for growth of tamoxifen-resistant cells (Butt
DpC and 4-OHT was synergistic in tamoxifen-sensitive and et al., 2005; Kilker et al., 2004; Kilker et al., 2006). Notably, cyclin D1
-resistant cells. and c-Myc are up-regulated by p-AKT, a major effector in the
To elucidate the mechanisms of synergy between DpC and PI3K/AKT pathway (Butt et al., 2005; Kilker et al., 2006). We found
4-OHT, major molecular effectors in the development of tamoxifen that DpC alone, or in combination with 4-OHT, strongly inhibited AKT
resistance were examined. ER-α was potently decreased by DpC in activation in sensitive and resistant MCF-7 cells. In fact, this inhibitory
sensitive and resistant MCF-7 cells using 2D and 3D culture. Further, effect on AKT may be responsible for the decreased cyclin D1 and c-
the combination of DpC with 4-OHT also decreased ER-α and Myc observed and could be central to the synergism between these
inhibited its transcriptional activity. These agents also decreased two two agents (Figure 6a,b).

F I G U R E 6 Diagram of the effects of


4-OHT alone or in combination with DpC
in MCF-7 tamoxifen-sensitive and
-resistant cells. (b) In the sensitive cells, E2
and 4-OHT compete to bind to the ER-α
ligand binding site. Binding of 4-OHT
results in conformational change of the
receptor, inhibiting its ability to promote
the transcription of genes containing
oestrogen response elements (EREs).
4-OHT also up-regulates p27, which
further inhibits the cell cycle. When
combined with 4-OHT, DpC inhibits p-
AKT, leading to decreased c-Myc and
cyclin D1 levels, which further decrease
tumour cell proliferation. (b) In tamoxifen-
resistant cells, increased activation of
non-genomic pathways enables enhanced
AKT activation, which leads to
(a) phosphorylation of the ER-α receptor
at Ser167, which enables its
transcriptional activity, despite 4-OHT
binding (Likhite et al., 2006); (b) activation
of downstream c-Myc and cyclin D1; and
(c) inhibition of p27 levels and activity
(Kilker et al., 2006; Motti et al., 2004).
Together, these effects drive proliferation
and form the major mechanisms of
resistance to 4-OHT. Combining DpC
with 4-OHT in these cells inhibits p-AKT,
leading to reduced phosphorylation of ER-
α receptor at Ser167, as well as
decreasing c-Myc and cyclin D1 levels,
consequently reducing tumour cell
proliferation. By inhibiting p-AKT, DpC
also enables increased expression of p27
to further inhibit cell cycle progression
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAQBOOL ET AL. 2377

p-AKT also promotes ER-α activation by Ser167 phosphorylation, resistance (Shee et al., 2018). FGF2 mediates its effects via the activa-
increasing ER-α binding to DNA and promoting its transcriptional tion of MEK/ERK signalling to promote cyclin D1, which is vital for
activity (Campbell et al., 2001; Likhite, Stossi, Kim, FGF2-mediated resistance (Shee et al., 2018). Notably, we demon-
Katzenellenbogen, & Katzenellenbogen, 2006). However, it is impor- strate here that DpC and particularly its combination with 4-OHT
tant to note that while high levels of p-ER-α (Ser167) can correlate to potently inhibited cyclin D1 expression. Hence, this therapeutic
good patient outcomes (Motomura et al., 2010), AKT is associated approach may potentially also inhibit TME-mediated mechanisms of
with poor outcomes and tamoxifen resistance in breast cancer resistance. However, further studies utilizing humanized in vivo
(Campbell et al., 2001). Here, DpC alone, or its combination with models that incorporate the TME are required to assess this.
4-OHT, markedly decreased p-ER-α (Ser167) in resistant MCF-7 cells. Overall, combining DpC with 4-OHT was more effective than
This may contribute to the synergy between DpC and 4-OHT, as the DpC alone against tamoxifen-resistant cells, as shown by (a) the
resistant cells can no longer activate ER-α via an intrinsic AKT- Chou–Talalay method demonstrating strong synergy between DpC
mediated mechanism, leaving them more vulnerable to 4-OHT. Alter- and 4-OHT (Figure 1c,f); (b) the combination treatment being most
natively, the ability of DpC to inhibit other major AKT targets effective at inhibiting ER-α transcriptional activity in resistant cells
(i.e., cyclin D1 and c-Myc) and ER-α co-activators (i.e., SRC3 and NF- (Figure 2c); (c) the combination treatment most potently reducing ER-
κB-p65) may be responsible for overcoming resistance to 4-OHT. α co-activators SRC3 and NF-κB-p65 in resistant cells (Figure S1); and
Another protein that mediates breast cancer endocrine resistance (d) only the combination treatment up-regulated the tumour suppres-
is the cyclin-dependent kinase inhibitor p27, with low levels being sor, p27, in resistant cells (Figure 4b). These results indicate that com-
associated with breast cancer relapse (Arteaga, 2004). Restoration of bining DpC with tamoxifen may be a promising therapeutic approach
p27 in endocrine-resistant breast cancer cells re-sensitizes them to for ER-positive breast cancer to overcome endocrine resistance.
anti-oestrogen therapies (Arteaga, 2004). We showed that, in sensi-
tive cells, 4-OHT up-regulated p27, which is in accordance with its AC KNOW LEDG EME NT S
anti-proliferative activity (Kilker et al., 2006; Figure 6a). However, in This project was supported by (a) Priority-driven Collaborative Cancer
resistant cells, p27 was decreased by 4-OHT, demonstrating signifi- Research Scheme (PdCCRS) Young Investigator Grant (1086449)
cant molecular alterations that confer tamoxifen resistance. While awarded to Z.K., which was co-funded by Cure Cancer Australia
DpC alone did not affect p27, its combination with 4-OHT induced a Foundation and Cancer Australia, and (b) PdCCRS Project Grant
significant increase in p27 in resistant cells. This synergy may be medi- (1146599) awarded to D.R.R. and Z.K. and co-funded by the National
ated by the ability of DpC to decrease p-AKT levels, as p-AKT pro- Breast Cancer Foundation and Cancer Australia. Z.K. is grateful for a
motes degradation of p27 (Kilker et al., 2006; Motti, De Marco, University of Sydney Bridging Fellowship, a National Health and Med-
Califano, Fusco, & Viglietto, 2004). ical Research Council RD Wright Fellowship (APP1140447), and a
The expression and activation of EGFR is another key difference Cancer Institute New South Wales (CINSW) Career Development Fel-
between tamoxifen-sensitive and -resistant cells, with this protein lowship (CDF171126). D.R.R. appreciates an NHMRC Senior Principal
being a major driver of tamoxifen resistance (Knowlden et al., 2003). Research Fellowship (1062607 and 1159596). P.J.J. was supported by
In agreement with this, we demonstrate that the resistant cells had a CINSW Career Development Fellowship (CDF141147). S.N.M
higher endogenous EGFR levels and responded robustly to E2 + EGF acknowledges funding provided by IRSIP-HEC (Pakistan) and the
treatment by increasing activating phosphorylation of EGFR, which Molecular Pathology and Pharmacology Program (USYD). We would
was in contrast to the sensitive cells. While DpC or its combination also like to thank Dr. Elizabeth Caldon (Garvan Institute of Medical
with 4-OHT increased EGFR expression and activation in the sensitive Research, Sydney) for providing the tamoxifen-sensitive and -resistant
cells, the activation of EGFR was reduced by DpC, and in particular its MCF-7 cells and valuable experimental advice.
combination with 4-OHT, in the resistant cells in the presence of
E2 + EGF. AUTHOR CONTRIBU TIONS
Importantly, downstream targets of EGFR signalling, namely, AKT, S.N.M. designed and performed experiments, analysed data, and
c-Myc, and cyclin D1, were all decreased by DpC and its combination wrote the manuscript. S.C.L. performed experiments and analysed
with 4-OHT in both sensitive and resistant cells. This suggests that data. R.H. analysed data. Z.K., P.J.J., and D.R.R. designed experiments,
increased EGFR levels/activation observed under some conditions analysed data, and wrote the manuscript.
may be a compensatory response to counteract the inhibition of
downstream signalling by DpC. Further, the inhibitory effect of these CONFLIC T OF INT ER E ST
agents on downstream AKT signalling may also be mediated by other The authors declare no conflicts of interest.
receptors, such as IGF1R and HER2, which were decreased by DpC
alone and in combination with 4-OHT in sensitive and resistant cells, DECLARATION OF T RANSPARENCY AND SCI ENTIFIC
respectively. RIGOUR
Another recently identified mechanism of anti-oestrogen resis- This Declaration acknowledges that this paper adheres to the princi-
tance involves the tumour micro-environment (TME), with FGF2 being ples for transparent reporting and scientific rigour of preclinical
produced by the TME and acting on breast cancer cells to promote research as stated in the BJP guidelines for Design & Analysis, and
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2378 MAQBOOL ET AL.

Immunoblotting and Immunochemistry, and as recommended by Chiarle, R., Pagano, M., & Inghirami, G. (2001). The cyclin dependent
funding agencies, publishers, and other organizations engaged with kinase inhibitor p27 and its prognostic role in breast cancer. Breast
Cancer Research, 3, 91–94. https://doi.org/10.1186/bcr277
supporting research.
Chou, T. C. (2006). Theoretical basis, experimental design, and computer-
ized simulation of synergism and antagonism in drug combination
ORCID studies. Pharmacological Reviews, 58, 621–681. https://doi.org/10.
Zaklina Kovacevic https://orcid.org/0000-0002-4742-4690 1124/pr.58.3.10
Chou, T. C. (2010). Drug combination studies and their synergy quantifica-
tion using the Chou-Talalay method. Cancer Research, 70, 440–446.
RE FE R ENC E S https://doi.org/10.1158/0008-5472.CAN-09-1947
Acconcia, F., & Kumar, R. (2006). Signaling regulation of genomic and Curtis, M. J., Alexander, S., Cirino, G., Docherty, J. R., George, C. H.,
nongenomic functions of estrogen receptors. Cancer Letters, Giembycz, M. A., … Ahluwalia, A. (2018). Experimental design and
238, 1–14. analysis and their reporting II: Updated and simplified guidance for
Alexander, S. P. H., Cidlowski, J. A., Kelly, E., Mathie, A., Peters, J. A., authors and peer reviewers. British Journal of Pharmacology, 175,
Veale, E. L., … CGTP Collaborators (2019). The Concise Guide to 987–993. https://doi.org/10.1111/bph.14153
PHARMACOLOGY 2019/20: Nuclear hormone receptors. British Jour- Edmondson, R., Broglie, J. J., Adcock, A. F., & Yang, L. (2014). Three-
nal of Pharmacology, 176, S229–S246. https://doi.org/10.1111/bph. dimensional cell culture systems and their applications in drug discov-
14750 ery and cell-based biosensors. Assay and Drug Development Technolo-
Alexander, S. P. H., Fabbro, D., Kelly, E., Mathie, A., Peters, J. A., gies, 12, 207–218. https://doi.org/10.1089/adt.2014.573
Veale, E. L., … CGTP Collaborators (2019a). The Concise Guide to Fan, P., Wang, J., Santen, R. J., & Yue, W. (2007). Long-term treatment
PHARMACOLOGY 2019/20: Catalytic receptors. British Journal of with tamoxifen facilitates translocation of estrogen receptor alpha out
Pharmacology, 176, S247–S296. https://doi.org/10.1111/bph.14751 of the nucleus and enhances its interaction with EGFR in MCF-7
Alexander, S. P. H., Fabbro, D., Kelly, E., Mathie, A., Peters, J. A., breast cancer cells. Cancer Research, 67, 1352–1360. https://doi.org/
Veale, E. L., … CGTP Collaborators (2019b). The Concise Guide to 10.1158/0008-5472.CAN-06-1020
PHARMACOLOGY 2019/20: Enzymes. British Journal of Pharmacol- Gao, J., & Richardson, D. R. (2001). The potential of iron chelators of the
ogy, 176, S297–S396. https://doi.org/10.1111/bph.14752 pyridoxal isonicotinoyl hydrazone class as effective antiproliferative
Ali, S., Rasool, M., Chaoudhry, H., Pushparaj, P. N., Jha, P., Hafiz, A., … agents, IV: The mechanisms involved in inhibiting cell-cycle progres-
Ali, A. (2016). Molecular mechanisms and mode of tamoxifen resis- sion. Blood, 98, 842–850. https://doi.org/10.1182/blood.v98.3.842
tance in breast cancer. Bioinformation, 12, 135–139. https://doi.org/ Ghayad, S. E., Vendrell, J. A., Ben Larbi, S., Dumontet, C., Bieche, I., &
10.6026/97320630012135 Cohen, P. A. (2010). Endocrine resistance associated with activated
Arteaga, C. L. (2004). Cdk inhibitor p27Kip1 and hormone dependence in ErbB system in breast cancer cells is reversed by inhibiting MAPK or
breast cancer. Clinical Cancer Research, 10, 368S–371S. https://doi. PI3K/Akt signaling pathways. International Journal of Cancer, 126,
org/10.1158/1078-0432.ccr-031204 545–562. https://doi.org/10.1002/ijc.24750
Benassayag, C., Montero, L., Colombie, N., Gallant, P., Cribbs, D., & Gionet, N., Jansson, D., Mader, S., & Pratt, M. A. (2009). NF-κB and estro-
Morello, D. (2005). Human c-Myc isoforms differentially regulate cell gen receptor α interactions: Differential function in estrogen receptor-
growth and apoptosis in Drosophila melanogaster. Molecular and Cellu- negative and -positive hormone-independent breast cancer cells. Jour-
lar Biology, 25, 9897–9909. https://doi.org/10.1128/MCB.25.22. nal of Cellular Biochemistry, 107, 448–459. https://doi.org/10.1002/
9897-9909.2005 jcb.22141
Biswas, D. K., Singh, S., Shi, Q., Pardee, A. B., & Iglehart, J. D. (2005). Green, A. R., Aleskandarany, M. A., Agarwal, D., Elsheikh, S., Nolan, C. C.,
Crossroads of estrogen receptor and NF-κB signaling. Science STKE Diez-Rodriguez, M., … Rakha, E. A. (2016). MYC functions are specific
2005: pe, 27. in biological subtypes of breast cancer and confers resistance to endo-
Bostner, J., Karlsson, E., Pandiyan, M. J., Westman, H., Skoog, L., crine therapy in luminal tumours. British Journal of Cancer, 114,
Fornander, T., … Stål, O. (2013). Activation of Akt, mTOR, and the 917–928. https://doi.org/10.1038/bjc.2016.46
estrogen receptor as a signature to predict tamoxifen treatment bene- Guo, Z. L., Richardson, D. R., Kalinowski, D. S., Kovacevic, Z., Tan-
fit. Breast Cancer Research and Treatment, 137, 397–406. https://doi. Un, K. C., & Chan, G. C. (2016). The novel thiosemicarbazone, di-
org/10.1007/s10549-012-2376-y 2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC),
Butt, A. J., McNeil, C. M., Musgrove, E. A., & Sutherland, R. L. (2005). inhibits neuroblastoma growth in vitro and in vivo via multiple mecha-
Downstream targets of growth factor and oestrogen signalling and nisms. Journal of Hematology & Oncology, 9, 98.
endocrine resistance: The potential roles of c-Myc, cyclin D1 and Hann, S. R. (2006). Role of post-translational modifications in regulating c-
cyclin E. Endocrine-Related Cancer, 12(Suppl 1), S47–S59. Myc proteolysis, transcriptional activity and biological function. Semi-
Campbell, R. A., Bhat-Nakshatri, P., Patel, N. M., Constantinidou, D., nars in Cancer Biology, 16, 288–302. https://doi.org/10.1016/j.
Ali, S., & Nakshatri, H. (2001). Phosphatidylinositol 3-kinase/AKT- semcancer.2006.08.004
mediated activation of estrogen receptor α: A new model for anti- Harding, S. D., Sharman, J. L., Faccenda, E., Southan, C., Pawson, A. J.,
estrogen resistance. The Journal of Biological Chemistry, 276, Ireland, S., … NC-IUPHAR (2018). The IUPHAR/BPS Guide to PHAR-
9817–9824. https://doi.org/10.1074/jbc.M010840200 MACOLOGY in 2018: Updates and expansion to encompass the new
Chang, M. (2012). Tamoxifen resistance in breast cancer. Biomol Ther guide to IMMUNOPHARMACOLOGY. Nucleic Acids Research, 46,
(Seoul), 20, 256–267. https://doi.org/10.4062/biomolther.2012.20. D1091–D1106. https://doi.org/10.1093/nar/gkx1121
3.256 Hayashi, S. I., Eguchi, H., Tanimoto, K., Yoshida, T., Omoto, Y., Inoue, A., …
Chen, Z., Zhang, D., Yue, F., Zheng, M., Kovacevic, Z., & Richardson, D. R. Yamaguchi, Y. (2003). The expression and function of estrogen recep-
(2012). The iron chelators Dp44mT and DFO inhibit TGF-β-induced tor alpha and beta in human breast cancer and its clinical application.
epithelial-mesenchymal transition via up-regulation of N-Myc Endocrine-Related Cancer, 10, 193–202. https://doi.org/10.1677/erc.
downstream-regulated gene 1 (NDRG1). The Journal of Biological 0.0100193
Chemistry, 287, 17016–17028. https://doi.org/10.1074/jbc.M112. Holliday, D. L., & Speirs, V. (2011). Choosing the right cell line for breast
350470 cancer research. Breast Cancer Research, 13, 215.
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
MAQBOOL ET AL. 2379

Jaiyesimi, I. A., Buzdar, A. U., Decker, D. A., & Hortobagyi, G. N. (1995). Lui, G. Y., Kovacevic, Z., Richardson, V., Merlot, A. M., Kalinowski, D. S., &
Use of tamoxifen for breast cancer: Twenty-eight years later. Journal Richardson, D. R. (2015). Targeting cancer by binding iron: Dissecting
of Clinical Oncology, 13, 513–529. https://doi.org/10.1200/JCO.1995. cellular signaling pathways. Oncotarget, 6, 18748–18779. https://doi.
13.2.513 org/10.18632/oncotarget.4349
Jansson, P. J., Kalinowski, D. S., Lane, D. J., Kovacevic, Z., Seebacher, N. A., Miller, M. A., & Katzenellenbogen, B. S. (1983). Characterization and quan-
Fouani, L., … Richardson, D. R. (2015). The renaissance of poly- titation of antiestrogen binding sites in estrogen receptor-positive and
pharmacology in the development of anti-cancer therapeutics: Inhibi- -negative human breast cancer cell lines. Cancer Research, 43,
tion of the “Triad of Death” in cancer by Di-2-pyridylketone 3094–3100.
thiosemicarbazones. Pharmacological Research, 100, 255–260. https:// Motomura, K., Ishitobi, M., Komoike, Y., Koyama, H., Nagase, H.,
doi.org/10.1016/j.phrs.2015.08.013 Inaji, H., & Noguchi, S. (2010). Expression of estrogen receptor beta
Kato, S., Endoh, H., Masuhiro, Y., Kitamoto, T., Uchiyama, S., Sasaki, H., … and phosphorylation of estrogen receptor alpha serine 167 correlate
Chambon, P. (1995). Activation of the estrogen receptor through with progression-free survival in patients with metastatic breast can-
phosphorylation by mitogen-activated protein kinase. Science, 270, cer treated with aromatase inhibitors. Oncology, 79, 55–61. https://
1491–1494. https://doi.org/10.1126/science.270.5241.1491 doi.org/10.1159/000319540
Kilker, R. L., Hartl, M. W., Rutherford, T. M., & Planas-Silva, M. D. (2004). Motti, M. L., De Marco, C., Califano, D., Fusco, A., & Viglietto, G. (2004).
Cyclin D1 expression is dependent on estrogen receptor function in Akt-dependent T198 phosphorylation of cyclin-dependent kinase
tamoxifen-resistant breast cancer cells. The Journal of Steroid Biochem- inhibitor p27kip1 in breast cancer. Cell Cycle, 3, 1074–1080.
istry and Molecular Biology, 92, 63–71. https://doi.org/10.1016/j. Nyga, A., Cheema, U., & Loizidou, M. (2011). 3D tumour models: Novel
jsbmb.2004.05.005 in vitro approaches to cancer studies. J Cell Commun Signal, 5,
Kilker, R. L., & Planas-Silva, M. D. (2006). Cyclin D1 is necessary for 239–248. https://doi.org/10.1007/s12079-011-0132-4
tamoxifen-induced cell cycle progression in human breast cancer cells. Obrero, M., Yu, D. V., & Shapiro, D. J. (2002). Estrogen receptor-
Cancer Research, 66, 11478–11484. https://doi.org/10.1158/0008- dependent and estrogen receptor-independent pathways for tamoxi-
5472.CAN-06-1755 fen and 4-hydroxytamoxifen-induced programmed cell death. The
Klinge, C. M. (2001). Estrogen receptor interaction with estrogen response Journal of Biological Chemistry, 277, 45695–45703. https://doi.org/10.
elements. Nucleic Acids Research, 29, 2905–2919. 1074/jbc.M208092200
Knowlden, J. M., Hutcheson, I. R., Jones, H. E., Madden, T., Gee, J. M., Osborne, C. K. (1998). Tamoxifen in the treatment of breast cancer. The
Harper, M. E., … Nicholson, R. I. (2003). Elevated levels of epidermal New England Journal of Medicine, 339, 1609–1618. https://doi.org/10.
growth factor receptor/c-erbB2 heterodimers mediate an autocrine 1056/NEJM199811263392207
growth regulatory pathway in tamoxifen-resistant MCF-7 cells. Endo- Osborne, C. K., Bardou, V., Hopp, T. A., Chamness, G. C., Hilsenbeck, S. G.,
crinology, 144, 1032–1044. https://doi.org/10.1210/en.2002-220620 Fuqua, S. A., … Schiff, R. (2003). Role of the estrogen receptor
Kovacevic, Z., Chikhani, S., Lovejoy, D. B., & Richardson, D. R. (2011). coactivator AIB1 (SRC-3) and HER-2/neu in tamoxifen resistance in
Novel thiosemicarbazone iron chelators induce up-regulation and breast cancer. Journal of the National Cancer Institute, 95, 353–361.
phosphorylation of the metastasis suppressor N-myc down-stream https://doi.org/10.1093/jnci/95.5.353
regulated gene 1: A new strategy for the treatment of pancreatic can- Osborne, C. K., Shou, J., Massarweh, S., & Schiff, R. (2005). Crosstalk
cer. Molecular Pharmacology, 80, 598–609. https://doi.org/10.1124/ between estrogen receptor and growth factor receptor pathways as a
mol.111.073627 cause for endocrine therapy resistance in breast cancer. Clinical Cancer
Kovacevic, Z., Menezes, S. V., Sahni, S., Kalinowski, D. S., Bae, D. H., Research, 11, 865s–870s.
Lane, D. J., & Richardson, D. R. (2016). The metastasis suppressor, N- Richardson, D. R., Tran, E. H., & Ponka, P. (1995). The potential of iron che-
MYC downstream-regulated gene-1 (NDRG1), down-regulates the lators of the pyridoxal isonicotinoyl hydrazone class as effective antip-
ErbB family of receptors to inhibit downstream oncogenic signaling roliferative agents. Blood, 86, 4295–4306.
pathways. The Journal of Biological Chemistry, 291, 1029–1052. Riggins, R. B., Schrecengost, R. S., Guerrero, M. S., & Bouton, A. H. (2007).
https://doi.org/10.1074/jbc.M115.689653 Pathways to tamoxifen resistance. Cancer Letters, 256, 1–24. https://
Lane, D. J., Saletta, F., Suryo Rahmanto, Y., Kovacevic, Z., & doi.org/10.1016/j.canlet.2007.03.016
Richardson, D. R. (2013). N-myc downstream regulated 1 (NDRG1) is Ring, A., & Dowsett, M. (2004). Mechanisms of tamoxifen resistance.
regulated by eukaryotic initiation factor 3a (eIF3a) during cellular Endocrine-Related Cancer, 11, 643–658. https://doi.org/10.1677/erc.
stress caused by iron depletion. PLoS ONE, 8, e57273. 1.00776
Lee, M. H., Koh, D., Na, H., Ka, N. L., Kim, S., Kim, H. J., … Lee, M. O. Santen, R. J., Fan, P., Zhang, Z., Bao, Y., Song, R. X., & Yue, W. (2009).
(2018). MTA1 is a novel regulator of autophagy that induces tamoxi- Estrogen signals via an extra-nuclear pathway involving IGF-1R and
fen resistance in breast cancer cells. Autophagy, 14, 812–824. https:// EGFR in tamoxifen-sensitive and -resistant breast cancer cells. Ste-
doi.org/10.1080/15548627.2017.1388476 roids, 74, 586–594. https://doi.org/10.1016/j.steroids.2008.11.020
Levin, E. R., & Pietras, R. J. (2008). Estrogen receptors outside the nucleus Sears, R. C. (2004). The life cycle of C-myc: From synthesis to degradation.
in breast cancer. Breast Cancer Research and Treatment, 108, 351–361. Cell Cycle, 3, 1133–1137.
https://doi.org/10.1007/s10549-007-9618-4 Sestak, V., Stariat, J., Cermanova, J., Potuckova, E., Chladek, J., Roh, J., …
Likhite, V. S., Stossi, F., Kim, K., Katzenellenbogen, B. S., & Kovarikova, P. (2015). Novel and potent anti-tumor and anti-
Katzenellenbogen, J. A. (2006). Kinase-specific phosphorylation of the metastatic di-2-pyridylketone thiosemicarbazones demonstrate mar-
estrogen receptor changes receptor interactions with ligand, ked differences in pharmacology between the first and second genera-
deoxyribonucleic acid, and coregulators associated with alterations in tion lead agents. Oncotarget, 6, 42411–42428. https://doi.org/10.
estrogen and tamoxifen activity. Molecular Endocrinology, 20, 18632/oncotarget.6389
3120–3132. https://doi.org/10.1210/me.2006-0068 Shee, K., Yang, W., Hinds, J. W., Hampsch, R. A., Varn, F. S.,
Lovejoy, D. B., Sharp, D. M., Seebacher, N., Obeidy, P., Prichard, T., Traphagen, N. A., … Miller, T. W. (2018). Therapeutically targeting
Stefani, C., … Richardson, D. R. (2012). Novel second-generation di- tumor microenvironment-mediated drug resistance in estrogen
2-pyridylketone thiosemicarbazones show synergism with standard receptor-positive breast cancer. The Journal of Experimental Medicine,
chemotherapeutics and demonstrate potent activity against lung can- 215, 895–910. https://doi.org/10.1084/jem.20171818
cer xenografts after oral and intravenous administration in vivo. Jour- Stabile, L. P., Lyker, J. S., Gubish, C. T., Zhang, W., Grandis, J. R., &
nal of Medicinal Chemistry, 55, 7230–7244. Siegfried, J. M. (2005). Combined targeting of the estrogen receptor
14765381, 2020, 10, Downloaded from https://bpspubs.onlinelibrary.wiley.com/doi/10.1111/bph.14985 by CAPES, Wiley Online Library on [24/02/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2380 MAQBOOL ET AL.

and the epidermal growth factor receptor in non-small cell lung cancer Yerushalmi, R., Woods, R., Ravdin, P. M., Hayes, M. M., & Gelmon, K. A.
shows enhanced antiproliferative effects. Cancer Research, 65, (2010). Ki67 in breast cancer: Prognostic and predictive potential. The
1459–1470. https://doi.org/10.1158/0008-5472.CAN-04-1872 Lancet Oncology, 11, 174–183. https://doi.org/10.1016/S1470-2045
Sun, M., Paciga, J. E., Feldman, R. I., Yuan, Z., Coppola, D., Lu, Y. Y., … (09)70262-1
Cheng, J. Q. (2001). Phosphatidylinositol-3-OH kinase (PI3K)/AKT2, Zhang, M. H., Man, H. T., Zhao, X. D., Dong, N., & Ma, S. L. (2014). Estro-
activated in breast cancer, regulates and is induced by estrogen recep- gen receptor-positive breast cancer molecular signatures and thera-
tor α (ERα) via interaction between ERα and PI3K. Cancer Research, 61, peutic potentials (Review). Biomed Rep, 2, 41–52. https://doi.org/10.
5985–5991. 3892/br.2013.187
Vranic, S., Gatalica, Z., & Wang, Z. Y. (2011). Update on the molecular pro-
file of the MDA-MB-453 cell line as a model for apocrine breast carci-
noma studies. Oncology Letters, 2, 1131–1137. https://doi.org/10. SUPPORTING INF ORMATION
3892/ol.2011.375 Additional supporting information may be found online in the
Xi, R., Pun, I. H., Menezes, S. V., Fouani, L., Kalinowski, D. S., Huang, M. L.,
Supporting Information section at the end of this article.
… Kovacevic, Z. (2017). Novel thiosemicarbazones inhibit lysine-rich
carcinoembryonic antigen-related cell adhesion molecule
1 (CEACAM1) coisolated (LYRIC) and the LYRIC-induced epithelial-
mesenchymal transition via upregulation of N-Myc downstream- How to cite this article: Maqbool SN, Lim SC, Park KC, et al.
regulated gene 1 (NDRG1). Molecular Pharmacology, 91, 499–517. Overcoming tamoxifen resistance in oestrogen receptor-
https://doi.org/10.1124/mol.116.107870 positive breast cancer using the novel thiosemicarbazone anti-
Yeh, E., Cunningham, M., Arnold, H., Chasse, D., Monteith, T., Ivaldi, G., …
cancer agent, DpC. Br J Pharmacol. 2020;177:2365–2380.
Sears, R. (2004). A signalling pathway controlling c-Myc degradation
that impacts oncogenic transformation of human cells. Nature Cell Biol- https://doi.org/10.1111/bph.14985
ogy, 6, 308–318. https://doi.org/10.1038/ncb1110

You might also like