You are on page 1of 10

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/340113064

Curcumin: a phytochemical modulator of estrogens and androgens in tumors


of the reproductive system

Article in Pharmacological Research · March 2020


DOI: 10.1016/j.phrs.2020.104765

CITATIONS READS

58 515

7 authors, including:

Mohammad Mohajeri Vanessa Bianconi


Mashhad University of Medical Sciences Università degli Studi di Perugia
28 PUBLICATIONS 1,070 CITATIONS 93 PUBLICATIONS 2,399 CITATIONS

SEE PROFILE SEE PROFILE

Marcos Avila Matteo Pirro


University of Tolima Università degli Studi di Perugia
89 PUBLICATIONS 2,231 CITATIONS 390 PUBLICATIONS 13,153 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

DYSfunctional HDL (DYS-HDL) study View project

COTININE AGAINST ALZHEIMERS DISEASE View project

All content following this page was uploaded by Mohammad Mohajeri on 16 March 2021.

The user has requested enhancement of the downloaded file.


Pharmacological Research 156 (2020) 104765

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Invited Review

Curcumin: a phytochemical modulator of estrogens and androgens in tumors T


of the reproductive system
Mohammad Mohajeria, Vanessa Bianconib, Marco Fidel Ávila-Rodriguezc, George E. Barretod,e,
Tannaz Jamialahmadif,g, Matteo Pirrob, Amirhossein Sahebkarh,i,j,*
a
Department of Medical Biotechnology & Nanotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
b
Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
c
Facultad de Ciencias de la Salud, Universidad del Tolima, Ibagué, Colombia
d
Department of Biological Sciences, University of Limerick, Limerick, Ireland
e
Health Research Institute, University of Limerick, Limerick, Ireland
f
Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
g
Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
h
Halal Research Center of IRI, FDA, Tehran, Iran
i
Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
j
School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran

ARTICLE INFO ABSTRACT

Keywords: Curcumin (Cur) is an active derivative extracted from turmeric which exerts a wide range of interactions with
curcumin biomolecules through complex signaling pathways. Cur has been extensively shown to possess potential anti-
hormonal activity tumor properties. In addition, there is growing body of evidence suggesting that Cur may exert potential anti-
androgen estrogen and anti-androgen activity. In vitro and in vivo studies suggest that anticancer properties of Cur against
estrogen
tumors affecting the reproductive system in females and males may be underlied by the Cur-mediated inhibition
cancer
of androgen and estrogen signaling pathways. In this review we examine various studies assessing the crosstalk
between Cur and both androgen and estrogen hormonal activity. Also, we discuss the potential chemopreventive
and antitumor role of Cur in the most prevalent cancers affecting the reproductive system in females and males.

1. Introduction type 2 diabetes, obesity, chronic inflammation, psychiatric and re-


spiratory diseases as well as some symptoms including pain and prur-
Curcumin (Cur; diferuloylmethane) is a natural polyphenol present itus [14–21]. In addition, there is growing evidence suggesting that Cur
in the turmeric (Curcuma longa) rhizomes [1–3]. Due to its broad may exert potential gonado-protective effects [22,23], due to its anti-
spectrum of biological properties [4–13], Cur has been widely re- oxidant [24], anti-inflammatory [25], anti-cancer [26,27], and anti-
cognized to exert potential therapeutic effects against a wide range of apoptotic activities, as well as its ability to modulate estrogen and an-
pathologies, including osteoarthritis, cardiac ischemia/reperfusion, drogen activities [28–32].
nonalcoholic fatty liver disease, metabolic syndrome, dyslipidaemia, Various in vitro and in vivo studies suggest that estrogens can

Abbreviations: AKR1C-2, aldoaldo-keto reductase family 1 member C2; Akt, protein kinase B; AP-1, activator protein-1; AR, androgen receptor; ASC-J9, di-
methylcurcumin; BC, breast cancer; b-FGF, basic fibroblast growth factor; CBP, cAMP response element-binding protein (CREB)-binding protein; CDK4, cyclin-
dependent kinase 4; CPM, Cur-loaded POCA4C6 micelles; CRPC, castrate-resistant PC; Cur, curcumin; CYP11A1, cytochrome P450, family 11, subfamily A, poly-
peptide 1; DCIS, ductal carcinoma in situ; DHT, dihydrotestosterone; EGCG, epigallocatechin gallate; EGFR, epidermal growth factor receptor; ER, estrogen receptor;
HC, hydrazinocurcumin; HER2, human epidermal growth factor; HSD3B2, 3 beta- and steroid delta-isomerase 2; ICOS, inducible T cell co-stimulator; IL, interleukin;
JAK, Janus kinase; MCP-1, monocyte chemoattractant protein-1; MHC, major histocompatibility complex; miR, microRNA; MMP-2, matrix metallopeptidase 2; NF-
kB, nuclear factor-KB; PAC, 4-hydroxy-3-methoxybenzylidene)-N-methyl-4-piperidone; PC, prostate cancer; PIAS, protein inhibitor of activated STAT; PLGA,
polylactide-co-glycolide; PSA, prostate-specific antigen; PSMA, prostate specific membrane antigen; RTKs, receptor tyrosine kinases; SAR, structure-activity re-
lationship; STAT3, signal transducer and activator of transcription 3; TIMP-1, tissue inhibitor metalloproteinase 1; TNBC, triple-negative breast cancer; TRAMP,
transgenic adenocarcinoma of the mouse prostate; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor

Corresponding author at: Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran.
E-mail addresses: sahebkara@mums.ac.ir, amir_saheb2000@yahoo.com (A. Sahebkar).

https://doi.org/10.1016/j.phrs.2020.104765
Received 2 July 2019; Received in revised form 14 February 2020; Accepted 18 March 2020
Available online 23 March 2020
1043-6618/ © 2020 Elsevier Ltd. All rights reserved.
M. Mohajeri, et al. Pharmacological Research 156 (2020) 104765

carcinogenesis and prostate cancer (PC) progression through the up-


regulation of AR and its target genes [41].
There is evidence showing that Cur and a number of Cur analogs
(Fig. 1) may exert chemopreventive and antitumor effects against
cancers of the reproductive system both in females and males, due to
their ability to modulate estrogen- and/or androgen-related pathways
[42–57]. In this review, we summarize the hormonal effects of Cur and
how they can interfere with estrogen- and androgen-dependent sig-
naling cascades in the most prevalent cancers affecting the reproductive
organs, that is BC, PC and endometrial cancer.

2. The chemopreventive and antitumor potential of curcumin


against tumors of the reproductive organs

Fig. 1. Chemical structure of curcumin and related compounds. There is growing body of evidence showing that anticancer effects
of Cur on the reproductive organs are related to the modulation of
increase the risk of endometrial and breast cancer (BC) by different androgen and/or estrogen-sensitive molecular targets involved in tu-
molecular mechanisms, including activation of cell growth, reduced morigenesis, cell proliferation and apoptosis. This section reviews
apoptosis, and DNA damage [33–40]. Also, androgens, beyond playing available literature concerning the role of estrogens and androgens in
a pivotal role in the normal prostate growth and functions through Cur-mediated anticancer activities against PC, BC and endometrial
various transcriptional cascades stimulated by binding of androgens to cancer.
the androgen receptor (AR), may also contribute to prostate

Fig. 2. Protective effects of curcumin on estrogenic tissues. Curcumin exerts protective effects on both estrogen receptor positive and negative tissues. Evidence
demonstrates the promissory actions of curcumin over endometrial carcinoma and breast cancer cell line models. The mechanism of action of curcumin in en-
dometrial carcinoma involves the diminished expression of growth factors receptors including FGF, HER, EGF, VEGF and PDGF causing reduced proliferation of
cancer cells. Additionally, curcumin exerts anti-proliferative and pro-apoptotic functions via Bax induction (Bax/Bcl-2 ratio > 1). Curcumin also induces Cyt-c
release, modulates P53 and promotes cell cycle arrest by regulation of ß-catenin/Wnt – Cyclin D1 signal pathways. In breast cancer models, curcumin diminishes ERα
expression and induces transcriptional modulation of ER expression via miR synthesis, i.e. miR-15a, miR-16, miR-344 and miR-22. The effects of curcumin also
extends to reduce the invasion and migration via the regulation of mediators such as laminin, MMP-2 and TIMP.

2
M. Mohajeri, et al. Pharmacological Research 156 (2020) 104765

Fig. 3. Effects of curcumin on androgenic tissues. Prostate tissue is a target of androgenic and estrogenic effectors via estrogen receptor beta (ERß), and androgen
receptor (AR). The induction of the prostatic tissue causes hypertrophy, enlargement and urethra obstruction. Chronic prostate (androgenic) stimulation may induce
prostate cancer (PC). Curcumin effects on PC cell models include reduction of cell proliferation, cell migration and androgen receptor expression. The molecular
mechanism involved the inhibition of AP-2γ and c-Jun. Curcumin also inhibits the activity of ß-catenin and NF-kB via Tcf-4, CBP and p300 proteins causing
diminished cell growth and migration. Interestingly, curcumin may also induce pro-apoptotic processes by promoting Cyt-c release and increasing Bax activity. It was
observed that curcumin diminishes PSA levels in cell models and clinical studies.

2.1. The chemopreventive and antitumor potential of curcumin against (0, 10, 25, and 50 μmol/L) on the suppression of androgen synthesis
prostate cancer were investigated in human PC cell lines (i.e., LNCaP and 22Rv1) [95].
It was found that Cur prevented cell growth and caused apoptosis of PC
Androgens are crucial for the control of prostate growth as well as cells in a dose-dependent manne by reducing the expression of ster-
its functions [44,58,59]. However, androgen signaling pathways are oidogenic acute regulatory proteins [i.e., cytochrome P450, family 11,
also implicated in PC formation and progression. Indeed, there is subfamily A, polypeptide 1 (CYP11A1) and 3 beta- and steroid delta-
growing body of evidence suggesting that PC progression tends to be isomerase 2 (HSD3B2)] and leading to the decline of testosterone
accompanied by an altered expression of AR coregulators (e.g., β-ca- synthesis. Cur treatment also caused a significant increase in the ex-
tenin) [60–67]. In addition, in some cases PCs become androgen-in- pression of aldo-keto reductase family 1 member C2 (AKR1C2), which
dependent by adjusting to a condition of low or no androgen supply was paralleled by a significant reduction of dihydrotestosterone (DHT).
through either a hyperactive AR or AR gene amplification [68–70]. In another study, the treatment of LNCaP cells with Cur (10 to 100 μM)
Noteworthy, intra-prostatic metabolism of androgens to estrogens was associated with a significant dose-dependent suppression of AR
through aromatase is important for the growth and function of prostate expression through the inhibition of the WnT/β-catenin pathway [96].
gland as well [71–76]. Indeed, estrogen signaling mediated through Also, in LNCaP and PC-3 cells Cur treatment (0-50 μM) down-regulated
estrogen receptor (ER)β negatively modulate prostate cell growth and the expression of AR and AR-related cofactors, including activator
migration ability [71,77–85]. protein-1 (AP-1), nuclear factor-KB (NF-kB), and cAMP response ele-
Among phytochemicals Cur has been widely studied as a chemo- ment-binding protein (CREB)-binding protein (CBP) [46]. In the same
preventive and antitumor agent against PC [86]. Available experi- study, Cur also inhibited the transforming activities of both cell lines as
mental studies suggest that Cur can modulate several androgen and/or evidenced by the declined colony-forming capacity in soft agar [46]. In
estrogen-dependent cell signaling pathways involved in the survival addition, in LNCaP PC cell lines, treatment with Cur (0, 5, 10, 20, 40,
and proliferation of PC cells [87–94]. To this regard, some in vitro and 80 μM) inhibited the androgen-mediated stimulation of prostate-
studies have demonstrated that Cur can down-regulate the expression specific antigen (PSA) gene expression through the down-regulation of
of both androgens and AR and inhibit the AR binding activity to the AR expression and activity [97]. In androgen-dependent LNCaP and
androgen-responsive element. In a study by Ide et al. the effects of Cur androgen-independent PC-3 cell lines, treatment with Cur (0, 10, 25,

3
M. Mohajeri, et al. Pharmacological Research 156 (2020) 104765

50, 75 and 100 μmol/L) [98] promoted the G2/M cell cycle arrest and antagonist, that is, exerted its function via competing with androgen
increased the ratios of apoptosis in a dose-dependent manner through binding to AR, it was found that [47] partial structures of DHT and ASC-
the down-regulation of AR expression. In a study by Banerjee et al. Cur J9 (A-ring of DHT and one phenyl ring of ASC-J9 as well as the hydroxy
(0, 5, 10, 15, 20, 25, and 50 μM) was added to reinforce the anti-tumor groups) were aligned. It was also shown that ASC-J9 and DHT possessed
properties of docetaxel against androgen-independent PC cells (i.e. the same conformations. The binding site of ASC-J9 on AR has not yet
DU145 and PC3) [89]. It was observed that the combined treatment had been recognized and whether it forms a bond to the androgen binding
a synergistic effect on the prevention of the proliferation and the in- site or how it interacts with AR is still unclear. Nonetheless, in several
duction of apoptosis of PC cell lines as compared to Cur or docetaxel experimental studies ASC-J9 has been observed to exert potent anti-PC
alone [89]. Noteworthy, the combined treatment reduced significantly effects by targeting either directly or indirectly the AR signaling
the expression of some receptor tyrosine kinases (RTKs), including the pathway [107]. Indeed, ASC-J9 was reported to inhibit CRPC cell
human epidermal growth factor (HER2) and the epidermal growth growth and metastasis by promoting the proteasome-mediated de-
factor receptor (EGFR), which are known to stimulate the AR signaling gradation of both wild-type AR and mutated AR (i.e., AR-F876 L)
by a ligand-independent mechanism in the androgen-independent stage [108–110]. It prevented tumor growth in mice with CWR-22Rv1 xe-
of advanced PC [89,99–101]. In androgen-sensitive LNCaP PC cell line nografts by degrading both full-length and splice variants of AR
and its bcl-2 over-expressing relative (LNCaP-bcl-2) it was found that [111,112]. It degraded ARs with the F876 L mutation in C4-2 and DU-
Cur (5 ± 50 μM) induced apoptosis by down-regulating the expression 145 cells and inhibited PC stem/progenitor cell invasion in mice with
of apoptosis suppressor proteins (i.e. bcl-2 or bcl-XL) as well as AR CWR-22Rv1 CD133 + S/P xenografts [110,113]. In LNCaP PC cells, it
protein [102]. Furthermore, in an in vitro study Cur was shown to in- was shown to selectively suppress AR expression by promoting the
hibit proliferation and migration of androgen-independent PC cell lines dissociation between AR and its coregulator ARA70 and leading to the
(i.e., DU145 and PC3) through estrogen-like effects, which were re- suppression of AR transactivation [114]. Overall, these findings suggest
versed following the administration of an anti-estrogen (i.e., ICI 182). that the anti-PC activity of ASC-J9 may be associated with the inhibi-
This suggested that Cur could potentially prevent the progression of tion of AR activity. However, it should be highlighted that ASC-J9 was
androgen-independent PC also by modulating ER action [44]. shown to inhibit cell growth of androgen-dependent LNCaP cells, even
The existence of Cur-mediated anti-androgen effects against PC has at a concentration of 1 μM, but not that of androgen-independent PC-3
been observed also in some in vivo studies. In prostate tissues from the or DU-145 cells at a concentration up to 5 μM [115]. This suggests that
transgenic adenocarcinoma of the mouse prostate (TRAMP) model a at higher doses ASC-J9 could prevent the proliferation of some AR-
four-week oral ingestion of Cur 200 mg/kg/d was associated with a negative PC cells through another functional mechanism than AR de-
significant decrease of testosterone and DHT levels, further suggesting gradation.
that Cur could possess potent anticancer features owing to the inhibi- Recently, numerous additional Cur analogs have been designed as
tion of androgen synthesis [95]. In an LNCaP xenograft model, Cur potential anti-PC drug candidates promoting anti-androgenic effects
500 mg/kg was shown to slow tumor growth by 27% during the early [45,47,115]. The structural variations in the novel analogs consist of
period by inhibiting AR activity [103,104]. In nude mice bearing substitutions around the biphenyl moiety, heteroaryl replacements of
LNCaP PC xenografts, a six-week synthetic diet containing 2% Cur re- one or both phenyl rings, alterations in the side chain(s) at the C4
duced tumor growth, by reducing the rate of PC cell growth and pro- methylene of the linker as well as modulation of the diketone or enol-
moting PC cell apoptosis. In addition, it was associated with a sig- ketone moiety and different lengths of the linker. SAR research showed
nificant reduction of microvessel density within excised tumors. that a suitable C4-side chain with the right size and physicochemical
Finally, some clinical studies have shown that Cur may have a ra- characteristics strongly determined their anti-AR activity. Compound 3
tional as an anti-androgen therapy against PC. In a phase II study en- with a propionic acid ethyl ester and compound 4 with an acrylic acid
rolling 30 patients with metastatic castrate-resistant PC (CRPC) without ethyl ester at the C4 methylene group exhibited a great potential not
any previous chemotherapy intervention, Cur (6,000 mg orally, once only to inhibit LNCap PC cell proliferation, but also to interfere with the
daily) in combination with docetaxel (75 mg/m [2] IV infusion, once transactivation of AR [45,115]. Interestingly compound 4 demon-
each three weeks for six cycles) induced an objective PSA response strated more potent anti-androgenic activity than compound 3. Com-
(decrease in the serum PSA of at least 50%) in 17 patients (59%) [105]. pounds 6–9, which are characterized by slight modifications in the C4-
However, the combined therapy with docetaxel and Cur in metastatic side chains as compared with compound 4, did not demonstrate any
CRPC [105] did not show any notable improvement in the overall specific anti-androgenic activity; nonetheless, they did reveal con-
survival. In a randomized clinical trial enrolling men who were found siderable cytotoxicity against both LNCap and PC-3 cells, suggesting
not to have PC in prostate biopsies, the combined effects of Cur (0, 2, that additional mechanisms beyond AR inhibition may explain their
20, and 50 μg/mL) and soy isoflavone (0, 0.1, 1, and 10 μg/mL) on antitumor activity. Compound 10, an analogue of compound 4 which
serum PSA levels were evaluated. The results showed that the PSA le- has free phenolic 4’-OH instead of 4’-OMe groups, failed to impede AR
vels were significantly reduced by the combined treatment among men activity in PC cells [116]. Thus, blockade of the phenolic 4’-OH groups
with pre-treatment serum PSA levels ≥ 10 μg/mL [106]. Nonetheless, is likely to promote an anti-androgenic activity. Substituting the two
despite this preliminary evidence on anti-androgenic effects of Cur in phenyls with other aryl rings or lengthening/shortening the length of
the clinical setting, Cur effectiveness as anti-cancer therapy against PC the linker connecting the two phenyls in Cur analogs was not found to
has yet to be fully examined. elevate their anti-androgenic activity, although different Cur analogs
with a shortened linker between two phenyls presented potent antic-
2.2. The chemopreventive and antitumor potential of curcumin analogs ancer effects [117]. Compound 11 including a 1,4-pentadiene-3-one
against prostate cancer moiety was shown to have a potent inhibitory action on LNCaP and PC-
3 cell growth, with an IC50 value of 0.5 ± 0.1 μM and 2.1 ± 1.1 μM,
Some Cur analogs have been shown to act as chemopreventive and respectively [118]. However, further investigations are warranted to
antitumor agents against PC by exerting anti-androgen effects. understand if this anti-androgenic activity may result from an increased
Particularly, different experimental studies have demonstrated that the AR degradation.
anticancer effects of some Cur analogs are mediated by the modulation Alterations in the enol-ketone moiety in the Cur structure led to
of AR signaling poor anti-androgenic activity, though the majority of these analogs
A novel Cur analog, dimethylcurcumin (ASC-J9), was identified displayed anticancer or cancer prevention features. Compound 12,
through structure-activity relationship (SAR) investigations [47]. In a where a 1H-pyrazole ring system substituted the enol-ketone moiety,
study aimed to elucidate if ASC-J9 acted like a conventional androgen unraveled more inhibitory potential against LNCaP cells (IC50:

4
M. Mohajeri, et al. Pharmacological Research 156 (2020) 104765

3.4 ± 0.9 μM) than PC-3 cells (IC50: 5.6 ± 2.0 μM) [118], suggesting environmental estrogens [43,143]. Cur (0-100 μM) was shown to pre-
that AR-mediated pathways may be responsible for these effects. vent the proliferation of T-47D BC cells in a dose-dependent manner
Whether this selective suppression in AR-positive tumor cells may be through a marked downregulation of ERα, ERβ and leptin [144], the
actually related to the anti-AR activity needs further research [107]. latter one being an adipocytokine with a crucial role in breast carci-
The effect of additional Cur analogs, including A10 ((2E,6E)-2,6-bis nogenesis regulated via ER-mediated transcription [144–147]. T-47D
(3,4,5-trimethoxybenzylidene) cyclohexanone), B10 ((2E,5E)-2,5-bis BC cells showed a significantly reduced expression of p53 and ERα
(3,4,5-trimethoxybenzylidene) cyclopentanone) and C10 ((1E,4E)-1,5- along with a reduced proliferation when treated with 20–80 μM Cur
bis(3,4,5-trimethoxyphenyl) penta-1,4-dien-3-one) without a het- [140]. The anti-proliferative properties of Cur were dependent on es-
eroatom linker, as well as E10 ((3E,5E)-3,5-bis (3,4,5-trimethox- trogens in ER-positive MCF-7 BC cells [43]. However, some in vitro
ybenzyli-dene) tetrahydropyran-4-one) and F10 ((3E,5E)-3,5-bis (3,4,5- studies suggested that the presence of estrogens was not necessary for
trimethoxybenzylidene) tetrahydrothiopyran-4-one) with a heteroatom Cur to exert anti-proliferative effects against BC. Indeed, Hallman et al.
linker were examined on human PC CWR-22Rv1 and LNCaP cell lines. showed that Cur (0, 5, 10, 20, 30, 40, 60, 80, 100 μM) reduced T-47D
Compounds F10 and E10 showed stronger inhibitory effects on the BC cell viability regardless the presence of anti-estrogens and ER ago-
viability of cultured CWR22Rv1 and LNCaP cell lines as compared with nists [140]. Shao et al. reported that Cur (0-50 μM) had suppressive
Cur and other Cur analogs. In addition, they tended to be more potent estrogen-independent effects on the proliferation and invasiveness of
inhibitors of AR activity than Cur, A10 and B10 [57]. ER-negative MDA-MB-231 BC cell lines [43]. Cur was shown to cause
The production of phase II metabolites (e.g., glucuronidation and cell cycle arrest in the G2/S and G2/M phase in a number of BC cell
sulfation) [119] as well as the poor bioavailability [120,121] make lines, including those resistant to anti-estrogens [148,149]. Overall,
unpredictable the pharmacokinetic of Cur in vivo. Piperine conjugation these findings suggest that additional mechanisms are involved in Cur-
has been used to prevent Cur hepatic and intestinal glucuronidation and mediated antitumor effects against BC beyond ER signaling pathways.
enhance its bioavailability [122]. Noteworthy, glucuronidation of xe- To this regard, it has recently been shown that Cur can inhibit BC on-
nobiotics is known to be possibly implicated in their conversions into cogenesis partly owing to the modulation of microRNA (miR)-mediated
carcinogens and to be involved in tumorigenesis [123]. Therefore, it is regulation of protein synthesis [150,151]. The expression of miR-15a,
conceivable that piperine-conjugated Cur antitumor effect may be miR-16 and miR-34a, which downregulate the expression of anti-
amplified. In addition, various nanoformulations based on liposomes, apoptotic proteins (e.g., Bcl-2 and Bmi-1), was shown to be increased in
cyclodextrins, polymers, and other special systems of delivery have different BC cell lines treated with Cur [152,153]. In MDA-MB-231 and
been used to improve the bioavailability of Cur [124]. Cur-based na- MDA-MB-435/β4 (β4-integrin transfectants) BC cell lines, Cur treat-
noparticle formulations can overcome the problems of poor oral ab- ment was shown to reduce cell motility and invasiveness by down-
sorption, probably through intravenous administration and therefore by regulating the expression of vascular endothelial growth factor (VEGF),
effectively modulating the exposure in plasma and promoting deposi- basic fibroblast growth factor (bFGF) and matrix metallopeptidase
tion into tumor tissue. In this regard, Yallapu et al. studied an anti- (MMP)-2, as well as by upregulating the expression of tissue inhibitor of
prostate specific membrane antigen (PSMA)-conjugated Cur-loaded metalloproteinase (TIMP-1) [154]. In BT-483 and MDA-MB-23 BC cell
poly(lactide-co-glycolide) (PLGA) nanoparticle with target selectivity lines, Cur was reported to inhibit BC cell proliferative rate and invasive
against PC cells manifesting PSMA in both in vitro and in vivo settings ability by down-regulating the expression of cyclin D1, cyclin-depen-
[125]. Lipid-polymer hybrid nanoparticles containing both docetaxel dent kinase 4 (CDK4) and MMP-1 [155]. Cur was also shown to induce
and Cur inhibited tumor growth in mice bearing PC-3 PC xenografts, cell cycle arrest at the G2M phase and late S phase in MCF-7 cells, by
implying the synergy of these two compounds [126]. A summary of suppressing the oncoprotein Aurora-A [156].
molecular structure of curcumin and related analogs/compounds is Some in vivo studies further support the notion that Cur may exert
displayed in Fig. 1. antitumor effects in BC by targeting both ER signaling and additional
molecular pathways. Lv et al. reported that Cur treatment led to a
2.3. The chemopreventive and antitumor potential of curcumin against significant reduction of tumor volumes and tumor weight in xenografts
breast cancer of MDA-MB-231 BC [157]. In athymic nude female mice implanted
with MDA-MB-231 cells, treatment with Cur (200 mg/kg/day, orally)
BC is the second main contributor of cancer death among women in plus epigallocatechin gallate (EGCG) (25 mg/kg/day, ip) for 10 weeks
the USA [127]. Although limited information is available on the asso- led to a 78% reduction in levels of VEGF receptor (VEGFR)-1 within the
ciation of circulating estrogen levels in premenopausal women with the tumors, which was paralleled by a 49% reduction in tumor volume with
risk of BC, an almost 2-fold higher risk of BC has been observed among combined treatment. Noteworthy, Cur treatment alone resulted also in
postmenopausal women in the top 20–25% (as opposed to bottom a marked reduction in tumor protein levels of EGFR and protein kinase
20–25%) of circulating estrogen levels [128]. Approximately 70% of B (Akt) [158].
BCs are categorized as ER-positive and can be treated using anti-es-
trogens [129,130]. Noteworthy, estrogens have a crucial pathogenic 2.4. The chemopreventive and antitumor potential of curcumin analogs
role also in the early intraductal neoplastic proliferation of epithelial against breast cancer
cells [i.e., ductal carcinoma in situ (DCIS)] preceding ER-positive in-
vasive BC [127,131,132]. Unlike ER-positive BC, triple-negative BC Some curcumin analogs have been shown to act as chemopreventive
(TNBC) is a heterogeneous and clinically aggressive disease that lacks and antitumor agents against BC by exerting anti-estrogen effects.
any approved targeted therapeutic approach as it possesses no receptor Hydrazinocurcumin (HC), a synthetic curcumin analogue, was shown to
for estrogen, progesterone, and human EGFR2 [133]. Since TNBC decrease the viability and migration ability of BC cell lines (i.e., MDA-
progression has been attributed to the up-regulation of β-catenin and MB-231, MCF-7) by inhibiting signal transducer and activator of tran-
AR, suppressing their expression or activity in BC stem cells can be scription 3 (STAT3) phosphorylation and downregulating STAT3
useful for the treatment of TNBC [134,135]. downstream targets, including Bcl-xL, Bcl-2, cyclin D1/D2, VEGF,
Among phytochemicals Cur has been widely studied as a chemo- MMP-2 and MMP-9 [159]. Encapsulation of ASC-J9 into PLGA nano-
preventive and antitumor agent against BC [136–142]. Several in vitro particles resulted in apoptosis of estrogen-dependent BC cells (MCF-7
studies have shown that Cur has inhibitory effects on the proliferation cells) at lower concentrations than the pure compound [141].
of human BC cells through the modulation of estrogen-induced ER- In view of Cur low bioavailability and low efficacy in the form of a
mediated transcription [43]. In ER-positive BC cells, potent anti-pro- free drug, a new vehicle for in vivo delivery of Cur using the phos-
liferative properties of Cur were reported in the presence of phorylated calixarene POCA4C6, was developed to assess the

5
M. Mohajeri, et al. Pharmacological Research 156 (2020) 104765

therapeutic potential of Cur against TNBC. Cur-loaded POCA4C6 mi- warrant further investigation in in vivo and clinical studies. A number of
celles (CPM) were developed by means of the thin-film method. The in vitro studies also suggest that Cur has multifaceted effects on an-
CPM consists of a unilamellar structure with a mean particle size of drogen signaling pathways in PC cells. Indeed, it was shown to promote
3.86 nm with high Cur encapsulation efficiency and loading as well as a the S5αR inhibition, induction of the cell cycle arrest in G2, the mod-
sustained release of Cur in a pH-dependent manner. This preparation ulation of Wnt/β-catenin signaling, the inhibition of PSA promoter
inhibited the proliferation, invasion, migration, and tumor spheroid activity, the suppression of NF-kB activation, the down-regulation of
formation of BT-549 and human BC cells. Increased apoptosis and re- apoptosis suppressor proteins and AR, the degradation of AR, and the
duced levels of nuclear β-catenin and AR were responsible for such block of the AR signaling pathway. However, the potential efficacy of
effects. Upon injection into tumor xenografts, CPM remained in the Cur in the treatment of PC needs to be further validated in clinical
tumor tissue and efficiently prevented its growth with no obvious sys- trials. Finally, growing experimental evidence showing that Cur may
temic toxicity. CPM also reduced the levels of CD44/CD133 BC stem exert significant antitumor effects against endometrial carcinoma needs
cells. These effects were hypothesized to be, at least in part, a result of to be corroborated by in vivo and clinical studies.
the disruption of AR-mediated suppressive signaling as well as of
crosstalk between AR signaling and Wnt/β-catenin signaling [160]. In a Declaration of Competing Interest
comparative study investigating the anticancer effects of Cur and its
analog 4-hydroxy-3-methoxybenzylidene)-N-methyl-4-piperidone None
(PAC), PAC was more stable than Cur in phosphate buffered saline and
in circulating blood. Differential sensitivities to PAC were found in ERα References
positive and ERα negative BC cells. Indeed, PAC totally inhibited ERα
only in ERα negative cells expressing a very low level of this receptor. [1] S. Jayakumar, R.S. Patwardhan, D. Pal, D. Sharma, S.K. Sandur,
The specific suppression of ERα in receptor-positive cells enhanced the Dimethoxycurcumin, a metabolically stable analogue of curcumin enhances the
radiosensitivity of cancer cells: Possible involvement of ROS and thioredoxin re-
apoptotic response to PAC, suggesting that ERα may prevent the PAC- ductase, Biochemical and biophysical research communications 478 (2016)
dependent induction of apoptosis via the ERα inhibition. In addition, 446–454.
PAC down-regulated the proliferation and reduced the epithelial-to- [2] R.S. Patwardhan, R. Checker, D. Sharma, V. Kohli, K.I. Priyadarsini, S.K. Sandur,
Dimethoxycurcumin, a metabolically stable analogue of curcumin, exhibits anti-
mesenchymal transition of BC cells both in vivo and in tumor xenografts. inflammatory activities in murine and human lymphocytes, Biochemical phar-
Finally, PAC inhibited the expression/release of two major cytokines macology 82 (2011) 642–657.
[i.e., interleukin (IL)-6 and monocyte chemoattractant protein-1 (MCP- [3] S.F. Soh, C.K. Huang, S.O. Lee, D. Xu, S. Yeh, J. Li, E.L. Yong, Y. Gong, C. Chang,
Determination of androgen receptor degradation enhancer ASC-J9((R)) in mouse
1), and prevented the paracrine procarcinogenic influences of BC cells
sera and organs with liquid chromatography tandem mass spectrometry, Journal
on breast stromal fibroblasts [161]. of pharmaceutical and biomedical analysis 88 (2014) 117–122.
[4] R.K. Maheshwari, A.K. Singh, J. Gaddipati, R.C. Srimal, Multiple biological ac-
tivities of curcumin: a short review, Life Sci 78 (2006) 2081–2087.
2.5. The chemopreventive and antitumor potential of curcumin/curcumin
[5] C. Martin-Cordero, M. Lopez-Lazaro, M. Galvez, M.J. Ayuso, Curcumin as a DNA
analogs against endometrial carcinoma topoisomerase II poison, J Enzyme Inhib Med Chem 18 (2003) 505–509.
[6] A.A. Momtazi, G. Derosa, P. Maffioli, M. Banach, A. Sahebkar, Role of microRNAs
In endometrial carcinoma cell lines (i.e., HEC-1-A cells), Cur ad- in the Therapeutic Effects of Curcumin in Non-Cancer Diseases, Mol Diagn Ther 20
(2016) 335–345.
ministration resulted in DNA degradation, apoptosis-like alterations [7] A.A. Momtaziand, A. Sahebkar, Difluorinated Curcumin: A Promising Curcumin
and decreased protein expression of the proto-oncogene Ets-1, as well Analogue with Improved Anti-Tumor Activity and Pharmacokinetic Profile, Curr
as the anti-apoptotic molecule Bcl-2, in a time and concentration-de- Pharm Des 22 (2016) 4386–4397.
[8] A. Mukhopadhyay, C. Bueso-Ramos, D. Chatterjee, P. Pantazis, B.B. Aggarwal,
pendent manner [162]. Also, in other endometrial carcinoma cell lines Curcumin downregulates cell survival mechanisms in human prostate cancer cell
(i.e., Ishikawa and RL95-2 cells) Cur inhibited cell growth and triggered lines, Oncogene 20 (2001) 7597–7609.
apoptosis through the inhibition of Janus kinase (JAK)-STAT signaling [9] C.H. Yeh, T.P. Chen, Y.C. Wu, Y.M. Lin, P. Jing Lin, Inhibition of NFkappaB ac-
tivation with curcumin attenuates plasma inflammatory cytokines surge and car-
via activation of the protein inhibitor of activated STAT (PIAS)-3 and diomyocytic apoptosis following cardiac ischemia/reperfusion, J Surg Res 125
attenuation of STAT3 phosphorylation [163]. In an in vivo study, Cur (2005) 109–116.
suppressed the mRNA and protein expression of Bcl-2 in endometrial [10] E. Abdollahi, A.A. Momtazi, T.P. Johnston, A. Sahebkar, Therapeutic effects of
curcumin in inflammatory and immune-mediated diseases: A nature-made jack-of-
carcinoma isolated from mice; however, co-administration of letrozole all-trades? Journal of Cellular Physiology 233 (2018) 830–848.
(an aromatase inhibitor) and Cur promoted tumor cell apoptosis [164]. [11] M.S. Karimian, M. Pirro, M. Majeed, A. Sahebkar, Curcumin as a natural regulator
Recently, the immunomodulatory potential of a Cur analog (i.e., Cur of monocyte chemoattractant protein-1, Cytokine and Growth Factor Reviews 33
(2017) 55–63.
phytosome) in endometrial carcinoma has been explored in a phase 2
[12] H. Mollazadeh, A.F.G. Cicero, C.N. Blesso, M. Pirro, M. Majeed, A. Sahebkar,
clinical study [165]. After consumption of 2 g/day Cur phytosome or- Immune modulation by curcumin: The role of interleukin-10, Critical Reviews in
ally for 2 weeks, blood samples from seven patients with endometrials Food Science and Nutrition 59 (2019) 89–101.
carcinoma showed a significant reduction of the expression levels of [13] A. Sahebkar, Molecular mechanisms for curcumin benefits against ischemic injury,
Fertility and Sterility 94 (2010) e75–e76.
major histocompatibility complex (MHC) on leukocytes and inducible T [14] Y. Panahi, M.S. Hosseini, N. Khalili, E. Naimi, L.E. Simental-Mendía, M. Majeed,
cell co-stimulator (ICOS) expression by CD8 + T cells, but no differ- A. Sahebkar, Effects of curcumin on serum cytokine concentrations in subjects
ences in inflammatory biomarkers, frequencies of immune cells other with metabolic syndrome: A post-hoc analysis of a randomized controlled trial,
Biomedicine & pharmacotherapy 82 (2016) 578–582.
than monocytes, and T cell activation. Therefore, whether Cur/Cur [15] Y. Panahi, R. Badeli, G.R. Karami, A. Sahebkar, Investigation of the efficacy of
analogues may represent possible immunoregulatory agents against adjunctive therapy with bioavailability‐boosted curcuminoids in major depressive
endometrial carcinoma remains uncertain [166]. disorder, Phytotherapy research 29 (2015) 17–21.
[16] D. Lelli, A. Sahebkar, T.P. Johnston, C. Pedone, Curcumin use in pulmonary dis-
eases: State of the art and future perspectives, Pharmacological research 115
3. Conclusions (2017) 133–148.
[17] A.A. Momtazi, G. Derosa, P. Maffioli, M. Banach, A. Sahebkar, Role of microRNAs
in the Therapeutic Effects of Curcumin in Non-Cancer Diseases, Molecular
Growing evidence indicates that Cur and its analogs interact with Diagnosis and Therapy 20 (2016) 335–345.
androgen and estrogen molecular pathways in different cell types and [18] Y. Panahi, P. Kianpour, R. Mohtashami, R. Jafari, L.E. Simental-Mendía,
conditions (Figs. 2 and 3). Various in vitro studies suggest that Cur and A. Sahebkar, Efficacy and Safety of Phytosomal Curcumin in Non-Alcoholic Fatty
Liver Disease: A Randomized Controlled Trial, Drug Research 67 (2017) 244–251.
its analogs can induce BC cell apoptosis and can inhibit the expression
[19] R. Rezaee, A.A. Momtazi, A. Monemi, A. Sahebkar, Curcumin: A potentially
of VEGFR-1, adipocytokines and cytokines by BC cells, by targeting powerful tool to reverse cisplatin-induced toxicity, Pharmacological Research 117
estrogen signaling pathways. These Cur-mediated effects, which may be (2017) 218–227.
potentially useful for the inhibition of BC growth and progression, [20] Y. Panahi, A. Sahebkar, M. Amiri, S.M. Davoudi, F. Beiraghdar, S.L. Hoseininejad,

6
M. Mohajeri, et al. Pharmacological Research 156 (2020) 104765

M. Kolivand, Improvement of sulphur mustard-induced chronic pruritus, quality of T. Chiang, E. Chang, Y. Lee, M.Y. Tsai, C. Chang, K.H. Lee, Antitumor agents. 217.
life and antioxidant status by curcumin: results of a randomised, double-blind, Curcumin analogues as novel androgen receptor antagonists with potential as anti-
placebo-controlled trial, British Journal of Nutrition 108 (2012) 1272–1279. prostate cancer agents, J Med Chem 45 (2002) 5037–5042.
[21] A. Sahebkarand, Y. Henrotin, Analgesic Efficacy and Safety of Curcuminoids in [48] P. Anand, C. Sundaram, S. Jhurani, A.B. Kunnumakkara, B.B. Aggarwal, Curcumin
Clinical Practice: A Systematic Review and Meta-Analysis of Randomized and cancer: an “old-age” disease with an “age-old” solution, Cancer letters 267
Controlled Trials, Pain Med 17 (2016) 1192–1202. (2008) 133–164.
[22] M. Azza, M. El-Wakf, M. Elhabiby, E. El-kholy, Use of tumeric and curcumin to [49] G. Saand T. Das, Anti cancer effects of curcumin: cycle of life and death, Cell
alleviate adverse reproductive outcomes of water: Nitrate pollution in male rats, division 3 (2008) 14.
Nat Sci 9 (2011) 229–239. [50] S. Singhand, A. Khar, Biological effects of curcumin and its role in cancer che-
[23] Y.O. Ilbey, E. Ozbek, M. Cekmen, A. Simsek, A. Otunctemur, A. Somay, Protective moprevention and therapy, Anti-Cancer Agents in Medicinal Chemistry (Formerly
effect of curcumin in cisplatin-induced oxidative injury in rat testis: mitogen-ac- Current Medicinal Chemistry-Anti-Cancer Agents) 6 (2006) 259–270.
tivated protein kinase and nuclear factor-kappa B signaling pathways, Human [51] D.L. Bemis, A.E. Katz, R. Buttyan, Clinical trials of natural products as chemo-
reproduction 24 (2009) 1717–1725. preventive agents for prostate cancer, Expert opinion on investigational drugs 15
[24] D.K. Sahoo, A. Roy, G.B. Chainy, Protective effects of vitamin E and curcumin on (2006) 1191–1200.
L-thyroxine-induced rat testicular oxidative stress, Chemico-biological interactions [52] S.C. Thomasset, D.P. Berry, G. Garcea, T. Marczylo, W.P. Steward, A.J. Gescher,
176 (2008) 121–128. Dietary polyphenolic phytochemicals—promising cancer chemopreventive agents
[25] E.O. Farombi, S.O. Abarikwu, I.A. Adedara, M.O. Oyeyemi, Curcumin and kola- in humans? A review of their clinical properties, International journal of cancer
viron ameliorate di‐n‐butylphthalate‐induced testicular damage in rats, Basic & 120 (2007) 451–458.
clinical pharmacology & toxicology 100 (2007) 43–48. [53] J. Hong, K. Ahn, E. Bae, S. Jeon, H. Choi, The effects of curcumin on the inva-
[26] A. Cort, M. Timur, E. Ozdemir, E. Kucuksayan, T. Ozben, Synergistic anticancer siveness of prostate cancer in vitro and in vivo, Prostate cancer and prostatic
activity of curcumin and bleomycin: an in vitro study using human malignant diseases 9 (2006) 147–152.
testicular germ cells, Mol Med Rep 5 (2012) 1481–1486. [54] T.O. Khor, Y.-S. Keum, W. Lin, J.-H. Kim, R. Hu, G. Shen, C. Xu,
[27] M. Iranshahi, A. Sahebkar, S.T. Hosseini, M. Takasaki, T. Konoshima, H. Tokuda, A. Gopalakrishnan, B. Reddy, X. Zheng, Combined inhibitory effects of curcumin
Cancer chemopreventive activity of diversin from Ferula diversivittata in vitro and and phenethyl isothiocyanate on the growth of human PC-3 prostate xenografts in
in vivo, Phytomedicine 17 (2010) 269–273. immunodeficient mice, Cancer Res. 66 (2006) 613–621.
[28] C. Aktas, M. Kanter, M. Erboga, S. Ozturk, Anti-apoptotic effects of curcumin on [55] J.-H. Kim, C. Xu, Y.-S. Keum, B. Reddy, A. Conney, A.-N.T. Kong, Inhibition of
cadmium-induced apoptosis in rat testes, Toxicology and industrial health 28 EGFR signaling in human prostate cancer PC-3 cells by combination treatment
(2012) 122–130. with β-phenylethyl isothiocyanate and curcumin, Carcinogenesis 27 (2005)
[29] A. Kadasi, N. Maruniakova, A. Stochmalova, M. Bauer, R. Grossmann, 475–482.
A.H. Harrath, A. Kolesarova, A.V. Sirotkin, Direct effect of curcumin on porcine [56] X. Wei, Z.-Y. Du, X. Zheng, X.-X. Cui, A.H. Conney, K. Zhang, Synthesis and eva-
ovarian cell functions, Animal reproduction science 182 (2017) 77–83. luation of curcumin-related compounds for anticancer activity, European journal
[30] H. Nurcahyoand, S.K. Soejono, . Effect of curcumin and pentagamavunon-0 on the of medicinal chemistry 53 (2012) 235–245.
steroidogenesis, proliferation activity and apoptosis in cultured porcine granulosa [57] D.Y. Zhou, N. Ding, Z.Y. Du, X.X. Cui, H. Wang, X.C. Wei, A.H. Conney, K. Zhang,
cells at varying stages of follicular growth, in: G.I.G. Pudjono Sismindari, X. Zheng, Curcumin analogues with high activity for inhibiting human prostate
S. Riyanto, E. Meiyanto, R.A. Susidarti, Ikawati, A.K. Nugroho, Ritmaleni (Eds.), cancer cell growth and androgen receptor activation, Mol Med Rep 10 (2014)
Recent development in curcumin pharmaco-chemistry, Proceedings of the 1315–1322.
International Symposium on Recent Progress in Curcumin Research, 11–12 [58] C.A. Heinleinand, C. Chang, Androgen receptor in prostate cancer, Endocrine re-
September 2006, Faculty of Pharmacy Gadjah Mada University, Yogyakarta, 2007, views 25 (2004) 276–308.
pp. 227–241. [59] E. Richter, S. Srivastava, A. Dobi, Androgen receptor and prostate cancer, Prostate
[31] J. Srivilai, K. Rabgay, N. Khorana, N. Waranuch, N. Nuengchamnong, Cancer Prostatic Dis 10 (2007) 114–118.
W. Wisuitiprot, T. Chuprajob, C. Changtam, A. Suksamrarn, W. Chavasiri, [60] M. Rahman, H. Miyamoto, C. Chang, Androgen receptor coregulators in prostate
N. Sornkaew, K. Ingkaninan, Anti-androgenic curcumin analogues as steroid 5- cancer: mechanisms and clinical implications, Clinical cancer research: an official
alpha reductase inhibitors, Medicinal Chemistry Research 26 (2017) 1550–1556. journal of the American Association for Cancer Research 10 (2004) 2208–2219.
[32] M. Mohamadpour, A. Noorafshan, S. Karbalay-Doust, T. Talaei-Khozani, [61] D.R. Chesireand, W.B. Isaacs, Beta-catenin signaling in prostate cancer: an early
E. Aliabadi, Protective effects of curcumin co-treatment in rats with establishing perspective, Endocrine-related cancer 10 (2003) 537–560.
chronic variable stress on testis and reproductive hormones, International journal [62] H. Clevers, Wnt/beta-catenin signaling in development and disease, Cell 127
of reproductive biomedicine (Yazd, Iran) 15 (2017) 447–452. (2006) 469–480.
[33] C.E. Caldon, Estrogen signaling and the DNA damage response in hormone de- [63] B.T. MacDonald, K. Tamai, X. He, Wnt/beta-catenin signaling: components, me-
pendent breast cancers, Frontiers in oncology 4 (2014) 106. chanisms, and diseases, Developmental cell 17 (2009) 9–26.
[34] B.E. Henderson, H.S. Feigelson, Hormonal carcinogenesis, Carcinogenesis 21 [64] D.J. Mulholland, H. Cheng, K. Reid, P.S. Rennie, C.C. Nelson, The androgen re-
(2000) 427–433. ceptor can promote beta-catenin nuclear translocation independently of adeno-
[35] S.-M. Ho, Estrogen, progesterone and epithelial ovarian cancer, Reproductive matous polyposis coli, The Journal of biological chemistry. 277 (2002)
Biology and Endocrinology 1 (2003) 73. 17933–17943.
[36] W. Yue, J.D. Yager, J.-P. Wang, E.R. Jupe, R.J. Santen, Estrogen receptor-depen- [65] F. Yang, X. Li, M. Sharma, C.Y. Sasaki, D.L. Longo, B. Lim, Z. Sun, Linking beta-
dent and independent mechanisms of breast cancer carcinogenesis, Steroids 78 catenin to androgen-signaling pathway, The Journal of biological chemistry 277
(2013) 161–170. (2002) 11336–11344.
[37] C.G.o.H.F.i.B, Cancer. Breast cancer and hormone replacement therapy: colla- [66] M. Verrasand, Z. Sun, Roles and regulation of Wnt signaling and beta-catenin in
borative reanalysis of data from 51 epidemiological studies of 52 705 women with prostate cancer, Cancer Lett 237 (2006) 22–32.
breast cancer and 108 411 women without breast cancer, The Lancet 350 (1997) [67] C.I. Truica, S. Byers, E.P. Gelmann, Beta-catenin affects androgen receptor tran-
1047–1059. scriptional activity and ligand specificity, Cancer Res 60 (2000) 4709–4713.
[38] C.G.o.H.F.i.B, Cancer. Menarche, menopause, and breast cancer risk: individual [68] A. Crocoll, C.C. Zhu, A.C. Cato, M. Blum, Expression of androgen receptor mRNA
participant meta-analysis, including 118 964 women with breast cancer from 117 during mouse embryogenesis, Mechanisms of development 72 (1998) 175–178.
epidemiological studies, The lancet oncology 13 (2012) 1141–1151. [69] P. Koivisto, J. Kononen, C. Palmberg, T. Tammela, E. Hyytinen, J. Isola,
[39] J. Hunnand, G.C. Rodriguez, Ovarian cancer: etiology, risk factors, and epide- J. Trapman, K. Cleutjens, A. Noordzij, T. Visakorpi, O.P. Kallioniemi, Androgen
miology, Clinical obstetrics and gynecology 55 (2012) 3–23. receptor gene amplification: a possible molecular mechanism for androgen de-
[40] V.W. Setiawan, H.P. Yang, M.C. Pike, S.E. McCann, H. Yu, Y.-B. Xiang, A. Wolk, privation therapy failure in prostate cancer, Cancer Res 57 (1997) 314–319.
N. Wentzensen, N.S. Weiss, P.M. Webb, Type I and II endometrial cancers: have [70] J. Kokontis, K. Takakura, N. Hay, S. Liao, Increased androgen receptor activity and
they different risk factors? Journal of Clinical Oncology 31 (2013) 2607–2618. altered c-myc expression in prostate cancer cells after long-term androgen depri-
[41] S.M. Green, E.A. Mostaghel, P.S. Nelson, Androgen action and metabolism in vation, Cancer Res 54 (1994) 1566–1573.
prostate cancer, Molecular and cellular endocrinology 360 (2012) 3–13. [71] G. Carruba, Estrogen and prostate cancer: An eclipsed truth in an androgen‐do-
[42] U. Banik, S. Parasuraman, A.K. Adhikary, N.H. Othman, Curcumin: the spicy minated scenario, Journal of cellular biochemistry 102 (2007) 899–911.
modulator of breast carcinogenesis, Journal of experimental & clinical cancer re- [72] S. Muthusamy, S. Andersson, H.-J. Kim, R. Butler, L. Waage, U. Bergerheim, J.-
search: CR 36 (98-98) (2017). Å. Gustafsson, Estrogen receptor β and 17β-hydroxysteroid dehydrogenase type 6,
[43] Z.M. Shao, Z.Z. Shen, C.H. Liu, M.R. Sartippour, V.L. Go, D. Heber, M. Nguyen, a growth regulatory pathway that is lost in prostate cancer, Proceedings of the
Curcumin exerts multiple suppressive effects on human breast carcinoma cells, Int National Academy of Sciences 108 (2011) 20090–20094.
J Cancer 98 (2002) 234–240. [73] M. Warnerand, J.-Å. Gustafsson, The role of estrogen receptor β (ERβ) in malig-
[44] M. Piccolella, V. Crippa, E. Messi, M.J. Tetel, A. Poletti, Modulators of estrogen nant diseases—A new potential target for antiproliferative drugs in prevention and
receptor inhibit proliferation and migration of prostate cancer cells, Pharmacol treatment of cancer, Biochemical and biophysical research communications 396
Res 79 (2014) 13–20. (2010) 63–66.
[45] L. Lin, Q. Shi, A.K. Nyarko, K.F. Bastow, C.C. Wu, C.Y. Su, C.C. Shih, K.H. Lee, [74] P. Negri‐Cesi, A. Colciago, A. Poletti, M. Motta, 5α‐reductase isozymes and ar-
Antitumor agents. 250. Design and synthesis of new curcumin analogues as po- omatase are differentially expressed and active in the androgen‐independent
tential anti-prostate cancer agents, J Med Chem 49 (2006) 3963–3972. human prostate cancer cell lines DU145 and PC3, The Prostate 41 (1999)
[46] K. Nakamura, Y. Yasunaga, T. Segawa, D. Ko, J.W. Moul, S. Srivastava, J.S. Rhim, 224–232.
Curcumin down-regulates AR gene expression and activation in prostate cancer [75] P. Negri‐Cesi, A. Poletti, A. Colciago, P. Magni, P. Martini, M. Motta, Presence of
cell lines, International journal of oncology 21 (2002) 825–830. 5α‐Reductase isozymes and aromatase in human prostate cancer cells and in be-
[47] H. Ohtsu, Z. Xiao, J. Ishida, M. Nagai, H.K. Wang, H. Itokawa, C.Y. Su, C. Shih, nign prostate hyperplastic tissue, The Prostate 34 (1998) 283–291.

7
M. Mohajeri, et al. Pharmacological Research 156 (2020) 104765

[76] A. Polettiand, L. Martini, Androgen-activating enzymes in the central nervous cancer-I. curcumin induces apoptosis in both androgen-dependent and androgen-
systemProceedings of Xth International Congress on Hormonal Steroids, Quebec, independent prostate cancer cells, Prostate Cancer Prostatic Dis 3 (2000) 84–93.
Canada, 17–21 June 1998, The Journal of steroid biochemistry and molecular [103] J.H. Hong, G. Lee, H.Y. Choi, Effect of curcumin on the interaction between an-
biology 69 (1999) 117–122. drogen receptor and Wnt/beta-catenin in LNCaP xenografts, Korean J Urol 56
[77] G. Kuiper, E. Enmark, M. Pelto-Huikko, S. Nilsson, J.-A. Gustafsson, Cloning of a (2015) 656–665.
novel receptor expressed in rat prostate and ovary, Proceedings of the National [104] B.C. Jordan, C.D. Mock, R. Thilagavathi, C. Selvam, Molecular mechanisms of
Academy of Sciences 93 (1996) 5925–5930. curcumin and its semisynthetic analogues in prostate cancer prevention and
[78] O.L.M. Oliveira, W.J. Koff, F. Muraro, E.B. Santos, D.F.G. Soares, V.M.T. Trindade, treatment, Life sciences 152 (2016) 135–144.
Steroid 5-α reductase type 2 activity in biopsies from malignant and normal [105] H. Mahammedi, E. Planchat, M. Pouget, X. Durando, H. Curé, L. Guy, I. Van-
prostatic tissues, Clinica Chimica Acta 391 (2008) 36–40. Praagh, L. Savareux, M. Atger, M. Bayet-Robert, The new combination docetaxel,
[79] H. Pettersson, L. Holmberg, M. Axelson, M. Norlin, CYP7B1‐mediated metabolism prednisone and curcumin in patients with castration-resistant prostate cancer: a
of dehydroepiandrosterone and 5α‐androstane‐3β, 17β‐diol–potential role (s) for pilot phase II study, Oncology 90 (2016) 69–78.
estrogen signaling, The FEBS journal 275 (2008) 1778–1789. [106] H. Ide, S. Tokiwa, K. Sakamaki, K. Nishio, S. Isotani, S. Muto, T. Hama, H. Masuda,
[80] G.S. Prinsand, K.S. Korach, The role of estrogens and estrogen receptors in normal S. Horie, Combined inhibitory effects of soy isoflavones and curcumin on the
prostate growth and disease, Steroids 73 (2008) 233–244. production of prostate-specific antigen, Prostate 70 (2010) 1127–1133.
[81] Z. Weihua, R. Lathe, M. Warner, J. Gustafsson, An endocrine pathway in the [107] Q. Shi, C.C. Shih, K.H. Lee, Novel anti-prostate cancer curcumin analogues that
prostate, ERbeta, AR, 5alpha-androstane-3beta, 17beta-diol, and CYP7B1, reg- enhance androgen receptor degradation activity, Anticancer Agents Med Chem 9
ulates prostate growth, Proceedings-National Academy of Sciences Usa 99 (2002) (2009) 904–912.
13589–13594. [108] D. Robinson, E.M. Van Allen, Y.-M. Wu, N. Schultz, R.J. Lonigro, J.-M. Mosquera,
[82] Z. Weihua, M. Warner, J.A. Gustafsson, Estrogen receptor beta in the prostate, Mol B. Montgomery, M.-E. Taplin, C.C. Pritchard, G. Attard, Integrative clinical
Cell Endocrinol 193 (2002) 1–5. genomics of advanced prostate cancer, Cell 161 (2015) 1215–1228.
[83] S. Steckelbroeck, Y. Jin, S. Gopishetty, B. Oyesanmi, T.M. Penning, [109] P.R. Dillard, M.F. Lin, S.A. Khan, Androgen-independent prostate cancer cells
Humancytosolic 3alpha-hydroxysteroid dehydrogenases of the aldo-keto re- acquire the complete steroidogenic potential of synthesizing testosterone from
ductasesuperfamily display significant 3beta-hydroxysteroid dehydrogenase ac- cholesterol, Mol Cell Endocrinol 295 (2008) 115–120.
tivity: implications for steroid hormone metabolism and action, Journal of bio- [110] R. Wang, W. Lin, C. Lin, L. Li, Y. Sun, C. Chang, ASC-J9((R)) suppresses castration
logical chemistry. 279 (2004) 10784–10795. resistant prostate cancer progression via degrading the enzalutamide-induced
[84] D. Dondi, M. Piccolella, A. Biserni, S. Della Torre, B. Ramachandran, A. Locatelli, androgen receptor mutant AR-F876L, Cancer Lett 379 (2016) 154–160.
P. Rusmini, D. Sau, D. Caruso, A. Maggi, Estrogen receptor β and the progression [111] T.-H. Lin, S.O. Lee, Y. Niu, D. Xu, L. Liang, L. Li, S.-D. Yeh, N. Fujimoto, S. Yeh,
of prostate cancer: role of 5α-androstane-3β, 17β-diol. Endocrine-related cancer C. Chang, Differential androgen deprivation therapies with anti-androgens ca-
17 (2010) 731–742. sodex/bicalutamide or MDV3100/enzalutamide versus anti-androgen receptor
[85] V. Guerini, D. Sau, P. Rusmini, P. Ciana, A. Maggi, P.G. Martini, ASC-J9® lead to promotion versus suppression of prostate cancer metastasis,
B.S. Katzenellenbogen, M. Motta, A. Poletti, The androgenderivative 5alpha-an- Journal of biological chemistry 288 (2013) 19359–19369.
drostane-3beta, 17beta-diol inhibits prostate cancer cellmigration through acti- [112] S. Yamashita, K.-P. Lai, K.-L. Chuang, D. Xu, H. Miyamoto, T. Tochigi, S.-T. Pang,
vation of the estrogen receptor beta subtype, Cancer Res 65 (2005) 5445–5453. L. Li, Y. Arai, H.-J. Kung, ASC-J9 suppresses castration-resistant prostate cancer
[86] J.G. Devassy, I.D. Nwachukwu, P.J. Jones, Curcumin and cancer: barriers to ob- growth through degradation of full-length and splice variant androgen receptors,
taining a health claim, Nutrition reviews 73 (2015) 155–165. Neoplasia 14 (2012) 74–83.
[87] B.B. Aggarwal, Prostate cancer and curcumin: add spice to your life, Cancer Biol [113] S. Wen, J. Tian, Y. Niu, L. Li, S. Yeh, C. Chang, ASC-J9®, and not Casodex or
Ther 7 (2008) 1436–1440. Enzalutamide, suppresses prostate cancer stem/progenitor cell invasion via al-
[88] R.A. Sharma, A.J. Gescher, W.P. Steward, Curcumin: the story so far, Eur J Cancer tering the EZH2-STAT3 signals, Cancer letters 376 (2016) 377–386.
41 (2005) 1955–1968. [114] Z. Yang, Y.J. Chang, I.C. Yu, S. Yeh, C.C. Wu, H. Miyamoto, D.E. Merry, G. Sobue,
[89] S. Banerjee, S.K. Singh, I. Chowdhury, J.W. Lillard Jr., R. Singh, Combinatorial L.M. Chen, S.S. Chang, C. Chang, ASC-J9 ameliorates spinal and bulbar muscular
effect of curcumin with docetaxel modulates apoptotic and cell survival molecules atrophy phenotype via degradation of androgen receptor, Nat Med 13 (2007)
in prostate cancer, Front Biosci 9 (2017) 235–245. 348–353.
[90] J. Li, S. Xiang, Q. Zhang, J. Wu, Q. Tang, J. Zhou, L. Yang, Z. Chen, S.S. Hann, [115] H. Ohtsu, H. Itokawa, Z. Xiao, C.Y. Su, C.C. Shih, T. Chiang, E. Chang, Y. Lee,
Combination of curcumin and bicalutamide enhanced the growth inhibition of S.Y. Chiu, C. Chang, K.H. Lee, Antitumor agents 222. Synthesis and anti-androgen
androgen-independent prostate cancer cells through SAPK/JNK and MEK/ERK1/ activity of new diarylheptanoids, Bioorg Med Chem. 11 (2003) 5083–5090.
2-mediated targeting NF-kappaB/p65 and MUC1-C, J Exp Clin Cancer Res 34 [116] L. Lin, Q. Shi, C.Y. Su, C.C. Shih, K.H. Lee, Antitumor agents 247. New 4-ethox-
(2015) 015–0168. ycarbonylethyl curcumin analogs as potential antiandrogenic agents, Bioorg Med
[91] V. Sharma, L. Kumar, S.K. Mohanty, J.P. Maikhuri, S. Rajender, G. Gupta, Chem 14 (2006) 2527–2534.
Sensitization of androgen refractory prostate cancer cells to anti-androgens [117] H. Ohori, H. Yamakoshi, M. Tomizawa, M. Shibuya, Y. Kakudo, A. Takahashi,
through re-expression of epigenetically repressed androgen receptor - Synergistic S. Takahashi, S. Kato, T. Suzuki, C. Ishioka, Y. Iwabuchi, H. Shibata, Synthesis and
action of quercetin and curcumin, Mol Cell Endocrinol 431 (2016) 12–23. biological analysis of new curcumin analogues bearing an enhanced potential for
[92] R. Wang, Y. Sun, L. Li, Y. Niu, W. Lin, C. Lin, E.S. Antonarakis, J. Luo, S. Yeh, the medicinal treatment of cancer, Molecular cancer therapeutics 5 (2006)
C. Chang, Preclinical Study using Malat1 Small Interfering RNA or Androgen 2563–2571.
Receptor Splicing Variant 7 Degradation Enhancer ASC-J9((R)) to Suppress [118] J.R. Fuchs, B. Pandit, D. Bhasin, J.P. Etter, N. Regan, D. Abdelhamid, C. Li, J. Lin,
Enzalutamide-resistant Prostate Cancer Progression, Eur Urol 72 (2017) 835–844. P.K. Li, Structure-activity relationship studies of curcumin analogues, Bioorganic
[93] M.M. Yallapu, S. Khan, D.M. Maher, M.C. Ebeling, V. Sundram, N. Chauhan, & medicinal chemistry letters 19 (2009) 2065–2069.
A. Ganju, S. Balakrishna, B.K. Gupta, N. Zafar, M. Jaggi, S.C. Chauhan, Anti-cancer [119] R.A. Sharma, H.R. McLelland, K.A. Hill, C.R. Ireson, S.A. Euden, M.M. Manson,
activity of curcumin loaded nanoparticles in prostate cancer, Biomaterials 35 M. Pirmohamed, L.J. Marnett, A.J. Gescher, W.P. Steward, Pharmacodynamic and
(2014) 8635–8648. pharmacokinetic study of oral Curcuma extract in patients with colorectal cancer,
[94] H. Ide, Y. Lu, R. Yamaguchi, S. Muto, S. Horie, Abstract 226: Chemopreventive Clinical cancer research: an official journal of the American Association for Cancer
potential of curcumin in prostate cancer, Cancer Res 74 (226-226) (2014). Research 7 (2001) 1894–1900.
[95] H. Ide, Y. Lu, T. Noguchi, S. Muto, H. Okada, S. Kawato, S. Horie, Modulation of [120] A.L. Cheng, C.H. Hsu, J.K. Lin, M.M. Hsu, Y.F. Ho, T.S. Shen, J.Y. Ko, J.T. Lin,
AKR1C2 by curcumin decreases testosterone production in prostate cancer, Cancer B.R. Lin, W. Ming-Shiang, H.S. Yu, S.H. Jee, G.S. Chen, T.M. Chen, C.A. Chen,
science 109 (2018) 1230–1238. M.K. Lai, Y.S. Pu, M.H. Pan, Y.J. Wang, C.C. Tsai, C.Y. Hsieh, Phase I clinical trial
[96] H. Choi, J. Lim, J. Hong, Curcumin interrupts the interaction between the an- of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant
drogen receptor and Wnt/β-catenin signaling pathway in LNCaP prostate cancer lesions, Anticancer Res 21 (2001) 2895–2900.
cells, Prostate cancer and prostatic diseases 13 (2010) 343. [121] N. Dhillon, B.B. Aggarwal, R.A. Newman, R.A. Wolff, A.B. Kunnumakkara,
[97] K.H. Tsui, T.H. Feng, C.M. Lin, P.L. Chang, H.H. Juang, Curcumin blocks the ac- J.L. Abbruzzese, C.S. Ng, V. Badmaev, R. Kurzrock, Phase II trial of curcumin in
tivation of androgen and interlukin-6 on prostate-specific antigen expression in patients with advanced pancreatic cancer, Clinical cancer research: an official
human prostatic carcinoma cells, J Androl 29 (2008) 661–668. journal of the American Association for Cancer Research 14 (2008) 4491–4499.
[98] H. Guo, Y.M. Xu, Z.Q. Ye, J.H. Yu, X.Y. Hu, Curcumin induces cell cycle arrest and [122] G. Shoba, D. Joy, T. Joseph, M. Majeed, R. Rajendran, P.S. Srinivas, Influence of
apoptosis of prostate cancer cells by regulating the expression of IkappaBalpha, c- piperine on the pharmacokinetics of curcumin in animals and human volunteers,
Jun and androgen receptor, Die Pharmazie 68 (2013) 431–434. Planta Med 64 (1998) 353–356.
[99] Z. Culig, A. Hobisch, M.V. Cronauer, C. Radmayr, J. Trapman, A. Hittmair, [123] J.Y. Li, F. Sun, H.F. Zhou, J. Yang, C. Huang, H. Fan, A Systematic Review
G. Bartsch, H. Klocker, Androgen receptor activation in prostatic tumor cell lines Exploring the Anticancer Activity and Mechanisms of Glucomannan, Frontiers in
by insulin-like growth factor-I, keratinocyte growth factor, and epidermal growth pharmacology 10 (2019) 930.
factor, Cancer research 54 (1994) 5474–5478. [124] M.M. Yallapu, P.K. Nagesh, M. Jaggi, S.C. Chauhan, Therapeutic Applications of
[100] S. Signoretti, R. Montironi, J. Manola, A. Altimari, C. Tam, G. Bubley, S. Balk, Curcumin Nanoformulations, Aaps J 17 (2015) 1341–1356.
G. Thomas, I. Kaplan, L. Hlatky, P. Hahnfeldt, P. Kantoff, M. Loda, Her-2-neu [125] M.M. Yallapu, S. Khan, D.M. Maher, M.C. Ebeling, V. Sundram, N. Chauhan,
expression and progression toward androgen independence in human prostate A. Ganju, S. Balakrishna, B.K. Gupta, N. Zafar, Anti-cancer activity of curcumin
cancer, Journal of the National Cancer Institute 92 (2000) 1918–1925. loaded nanoparticles in prostate cancer, Biomaterials 35 (2014) 8635–8648.
[101] J. Andersson, M. Rosestedt, V. Asplund, N. Yavari, A. Orlova, In vitro modeling of [126] J. Yan, Y. Wang, X. Zhang, S. Liu, C. Tian, H. Wang, Targeted nanomedicine for
HER2-targeting therapy in disseminated prostate cancer, International journal of prostate cancer therapy: docetaxel and curcumin co-encapsulated lipid–polymer
oncology 45 (2014) 2153–2158. hybrid nanoparticles for the enhanced anti-tumor activity in vitro and in vivo,
[102] T. Dorai, N. Gehani, A. Katz, Therapeutic potential of curcumin in human prostate Drug delivery 23 (2016) 1757–1762.

8
M. Mohajeri, et al. Pharmacological Research 156 (2020) 104765

[127] L. Wiechmannand, H.M. Kuerer, The molecular journey from ductal carcinoma in [147] J.S. O’Neil, M.E. Burow, A.E. Green, J.A. McLachlan, M.C. Henson, Effects of es-
situ to invasive breast cancer, Cancer 112 (2008) 2130–2142. trogen on leptin gene promoter activation in MCF-7 breast cancer and JEG-3
[128] S.B. Brownand, S.E. Hankinson, Endogenous estrogens and the risk of breast, en- choriocarcinoma cells: selective regulation via estrogen receptors α and β,
dometrial, and ovarian cancers, Steroids 99 (2015) 8–10. Molecular and cellular endocrinology 176 (2001) 67–75.
[129] W.F. Anderson, N. Chatterjee, W.B. Ershler, O.W. Brawley, Estrogen receptor [148] M. Jiang, O. Huang, X. Zhang, Z. Xie, A. Shen, H. Liu, M. Geng, K. Shen, Curcumin
breast cancer phenotypes in the Surveillance, Epidemiology, and End Results da- induces cell death and restores tamoxifen sensitivity in the antiestrogen-resistant
tabase, Breast cancer research and treatment 76 (2002) 27–36. breast cancer cell lines MCF-7/LCC2 and MCF-7/LCC9, Molecules (Basel,
[130] T. Sorlie, C.M. Perou, R. Tibshirani, T. Aas, S. Geisler, H. Johnsen, T. Hastie, Switzerland) 18 (2013) 701–720.
M.B. Eisen, M. van de Rijn, S.S. Jeffrey, T. Thorsen, H. Quist, J.C. Matese, [149] K. Mehta, P. Pantazis, T. McQueen, B.B. Aggarwal, Antiproliferative effect of
P.O. Brown, D. Botstein, P.E. Lonning, A.L. Borresen-Dale, Gene expression pat- curcumin (diferuloylmethane) against human breast tumor cell lines, Anti-cancer
terns of breast carcinomas distinguish tumor subclasses with clinical implications, drugs 8 (1997) 470–481.
Proceedings of the National Academy of Sciences of the United States of America [150] S. Norouzi, M. Majeed, M. Pirro, D. Generali, A. Sahebkar, Curcumin as an Adjunct
98 (2001) 10869–10874. Therapy and microRNA Modulator in Breast Cancer, Curr Pharm Des 24 (2018)
[131] Y. Vaidya, P. Vaidya, T. Vaidya, Ductal carcinoma in situ of the breast, Indian 171–177.
Journal of Surgery 77 (2015) 141–146. [151] A.A. Momtazi, F. Shahabipour, S. Khatibi, T.P. Johnston, M. Pirro, A. Sahebkar,
[132] C.F. Cowell, B. Weigelt, R.A. Sakr, C.K. Ng, J. Hicks, T.A. King, J.S. Reis-Filho, Curcumin as a MicroRNA Regulator in Cancer: A Review. Reviews of physiology,
Progression from ductal carcinoma in situ to invasive breast cancer: revisited, biochemistry and pharmacology 171 (2016) 1–38.
Molecular oncology 7 (2013) 859–869. [152] J. Guo, W. Li, H. Shi, X. Xie, L. Li, H. Tang, M. Wu, Y. Kong, L. Yang, J. Gao, P. Liu,
[133] N. Aceto, N. Sausgruber, H. Brinkhaus, D. Gaidatzis, G. Martiny-Baron, W. Wei, X. Xie, Synergistic effects of curcumin with emodin against the pro-
G. Mazzarol, S. Confalonieri, M. Quarto, G. Hu, P.J. Balwierz, M. Pachkov, liferation and invasion of breast cancer cells through upregulation of miR-34a,
S.J. Elledge, E. van Nimwegen, M.B. Stadler, M. Bentires-Alj, Tyrosine phosphatase Molecular and cellular biochemistry 382 (2013) 103–111.
SHP2 promotes breast cancer progression and maintains tumor-initiating cells via [153] J. Yang, Y. Cao, J. Sun, Y. Zhang, Curcumin reduces the expression of Bcl-2 by
activation of key transcription factors and a positive feedback signaling loop, Nat upregulating miR-15a and miR-16 in MCF-7 cells, Medical oncology (Northwood,
Med 18 (2012) 529–537. London, England) 27 (2010) 1114–1118.
[134] N. Dey, B.G. Barwick, C.S. Moreno, M. Ordanic-Kodani, Z. Chen, G. Oprea-Ilies, [154] H.I. Kim, H. Huang, S. Cheepala, S. Huang, J. Chung, Curcumin inhibition of in-
W. Tang, C. Catzavelos, K.F. Kerstann, G.W. Sledge Jr., M. Abramovitz, M. Bouzyk, tegrin (alpha6beta4)-dependent breast cancer cell motility and invasion, Cancer
P. De, B.R. Leyland-Jones, Wnt signaling in triple negative breast cancer is asso- prevention research (Philadelphia, Pa) 1 (2008) 385–391.
ciated with metastasis, BMC cancer 13 (2013) 537. [155] Q. Liu, W.T. Loo, S.C. Sze, Y. Tong, Curcumin inhibits cell proliferation of MDA-
[135] T. Proverbs-Singh, J.L. Feldman, M.J. Morris, K.A. Autio, T.A. Traina, Targeting MB-231 and BT-483 breast cancer cells mediated by down-regulation of
the androgen receptor in prostate and breast cancer: several new agents in de- NFkappaB, cyclinD and MMP-1 transcription, Phytomedicine 16 (2009) 916–922.
velopment, Endocrine-related cancer 22 (R87-r106) (2015). [156] C.S. Ke, H.S. Liu, C.H. Yen, G.C. Huang, H.C. Cheng, C.Y. Huang, C.L. Su,
[136] M.M. Manson, P.B. Farmer, A. Gescher, W.P. Steward, Innovative agents in cancer Curcumin-induced Aurora-A suppression not only causes mitotic defect and cell
prevention, Recent Results Cancer Res 166 (2005) 257–275. cycle arrest but also alters chemosensitivity to anticancer drugs, J Nutr Biochem
[137] M.S. Squires, E.A. Hudson, L. Howells, S. Sale, C.E. Houghton, J.L. Jones, L.H. Fox, 25 (2014) 526–539.
M. Dickens, S.A. Prigent, M.M. Manson, Relevance of mitogen activated protein [157] Z.D. Lv, X.P. Liu, W.J. Zhao, Q. Dong, F.N. Li, H.B. Wang, B. Kong, Curcumin
kinase (MAPK) and phosphotidylinositol-3-kinase/protein kinase B (PI3K/PKB) induces apoptosis in breast cancer cells and inhibits tumor growth in vitro and in
pathways to induction of apoptosis by curcumin in breast cells, Biochemical vivo, Int J Clin Exp Pathol 7 (2014) 2818–2824.
pharmacology 65 (2003) 361–376. [158] T.J. Somers-Edgar, M.J. Scandlyn, E.C. Stuart, M.J. Le Nedelec, S.P. Valentine,
[138] G.M. Calaf, C. Echiburu-Chau, G. Wen, A.S. Balajee, D. Roy, Effect of curcumin on R.J. Rosengren, The combination of epigallocatechin gallate and curcumin sup-
irradiated and estrogen-transformed human breast cell lines, International journal presses ER alpha-breast cancer cell growth in vitro and in vivo, Int J Cancer 122
of oncology 40 (2012) 436–442. (2008) 1966–1971.
[139] M. de Freitas Silva, L.F. Coelho, I.M. Guirelli, R.M. Pereira, G.A. Ferreira-Silva, [159] X. Wang, Y. Zhang, X. Zhang, W. Tian, W. Feng, T. Chen, The curcumin analogue
G.Y. Graravelli, R.O. Horvath, E.S. Caixeta, M. Ionta, C. Viegas, Synthetic re- hydrazinocurcumin exhibits potent suppressive activity on carcinogenicity of
sveratrol-curcumin hybrid derivative inhibits mitosis progression in estrogen po- breast cancer cells via STAT3 inhibition, International journal of oncology. 40
sitive MCF-7 breast cancer cells, Toxicol In Vitro 50 (2018) 75–85. (2012) 1189–1195.
[140] K. Hallman, K. Aleck, B. Dwyer, V. Lloyd, M. Quigley, N. Sitto, A.E. Siebert, [160] W. Chen, L. Li, X. Zhang, Y. Liang, Z. Pu, L. Wang, J. Mo, Curcumin: a calixarene
S. Dinda, The effects of turmeric (curcumin) on tumor suppressor protein (p53) derivative micelle potentiates anti-breast cancer stem cells effects in xenografted,
and estrogen receptor (ERα) in breast cancer cells, Breast Cancer: Targets and triple-negative breast cancer mouse models, Drug Deliv 24 (2017) 1470–1481.
Therapy 9 (2017) 153–161. [161] H.A. Al-Howail, H.A. Hakami, B. Al-Otaibi, A. Al-Mazrou, M.H. Daghestani, I. Al-
[141] P. Verderio, L. Pandolfi, S. Mazzucchelli, M.R. Marinozzi, R. Vanna, F. Gramatica, Jammaz, H.H. Al-Khalaf, A. Aboussekhra, PAC down-regulates estrogen receptor
F. Corsi, M. Colombo, C. Morasso, D. Prosperi, Antiproliferative effect of ASC-J9 alpha and suppresses epithelial-to-mesenchymal transition in breast cancer cells,
delivered by PLGA nanoparticles against estrogen-dependent breast cancer cells, BMC cancer 16 (2016) 540.
Mol Pharm 11 (2014) 2864–2875. [162] Z. Yuand, D.M. Shah, Curcumin down-regulates Ets-1 and Bcl-2 expression in
[142] S.P. Verma, E. Salamone, B. Goldin, Curcumin and genistein, plant natural pro- human endometrial carcinoma HEC-1-A cells, Gynecologic oncology 106 (2007)
ducts, show synergistic inhibitory effects on the growth of human breast cancer 541–548.
MCF-7 cells induced by estrogenic pesticides, Biochemical and biophysical re- [163] M. Saydmohammed, D. Joseph, V. Syed, Curcumin suppresses constitutive acti-
search communications 233 (1997) 692–696. vation of STAT‐3 by up‐regulating protein inhibitor of activated STAT‐3 (PIAS‐3)
[143] S.P. Verma, B.R. Goldin, P.S. Lin, The inhibition of the estrogenic effects of pes- in ovarian and endometrial cancer cells, Journal of cellular biochemistry 110
ticides and environmental chemicals by curcumin and isoflavonoids, (2010) 447–456.
Environmental health perspectives 106 (1998) 807–812. [164] Y.-J. Liang, Q. Hao, Y.-Z. Wu, Q.-L. Wang, J.-D. Wang, Y.-L. Hu, Aromatase in-
[144] K. Nejati-Koshki, A. Akbarzadeh, M. Pourhassan-Moghaddam, Curcumin inhibits hibitor letrozole in synergy with curcumin in the inhibition of xenografted en-
leptin gene expression and secretion in breast cancer cells by estrogen receptors, dometrial carcinoma growth, International Journal of Gynecological Cancer 19
Cancer cell international 14 (2014) 66. (2009) 1248–1252.
[145] L. Vona-Davisand, D.P. Rose, Adipokines as endocrine, paracrine, and autocrine [165] S. Tuyaerts, K. Rombauts, T. Everaert, A.M.T. Van Nuffel, F. Amant, A Phase 2
factors in breast cancer risk and progression, Endocrine-related cancer 14 (2007) Study to Assess the Immunomodulatory Capacity of a Lecithin-based Delivery
189–206. System of Curcumin in Endometrial Cancer, Frontiers in nutrition 5 (2018) 138.
[146] K.W. Yi, J.H. Shin, H.S. Seo, J.K. Lee, M.J. Oh, T. Kim, H.S. Saw, S.H. Kim, [166] A. Bahrami, M. Fereidouni, M. Pirro, V. Bianconi, A. Sahebkar, Modulation of
J.Y. Hur, Role of Estrogen Receptor‐α and− β in Regulating Leptin Expression in regulatory T cells by natural products in cancer, Cancer Lett 459 (2019) 72–85.
3T3‐L1 Adipocytes, Obesity 16 (2008) 2393–2399.

View publication stats

You might also like